Abstract
The strength of the interaction between T cell receptors (TCRs) and their ligands, peptide/MHC complexes (pMHCs), is one of the most frequently discussed and investigated features of T cells in immuno-oncology today. Although there are many molecules on the surface of T cells that interact with ligands on other cells, the TCR/pMHC is the only receptor-ligand pair that offers antigen specificity and dictates the functional response of the T cell. The strength of the TCR/pMHC interaction, along with the environment in which this interaction takes place, is key to how the T cell will respond. The TCR repertoire of T cells that interact with tumor-associated antigens is vast, although typically of low affinity. Here we focus on the low affinity interactions between TCRs from CD8+ T cells and different models used in immuno-oncology.
Keywords: TCR-pMHC affinity, Tumor antigens, CD8+ T cells, Altered peptide ligands, Low-affinity
Introduction
The binding strength of T cell receptors (TCRs) on CD8+ T cells to specific MHC class I molecules complexed with self peptides (a.k.a. tumor-associated antigens, TAAs) is generally weak1. On average, TCRs bind to these self pMHC molecules at 10 times lower binding affinity than to foreign antigens2. These naturally selected self antigen TCR affinities are in the range of 1–100 μM3. Many tumor antigens are, in fact, self antigens that have become dysregulated during the transformation process4–6. Frequently, these tumor-associated antigens (TAAs) are much more highly expressed in the tumor than the periphery and can be shared among different tumors. For example, expression of the prototype TAA NY-ESO-1 is only found in the immune-privileged testes, but is expressed in many different kinds of tumors7. The shared expression of TAAs, in opposition to neoantigens, makes them attractive targets for immunotherapy. In addition to TCRs binding to many peptide-MHC molecules on an antigen presenting cell, numerous interactions between molecules other than TCR-pMHC contribute to the avidity of the interaction. However, in this review we focus primarily on the TCR-pMHC interaction and refer to the strength of this binding as the “affinity.”
T cell fate is determined by TCR engagement and downstream signaling in both the thymus and periphery. To avoid autoimmune responses, but protect from pathogens, T cells in the periphery must be tolerized against self, yet strongly recognize foreign pathogens. In the thymus, the transcription factor AIRE aides expression of self antigens on thymic epithelial cells, facilitating their presentation to developing T cells8. Strong interactions lead to negative selection and deletion of self-reactive T cells, or alternatively, T cell differentiation into regulatory T cells9,10. The KD of ~6 μM, just about the negative selection threshold, is termed the “danger zone” for autoantigens, as negative selection functions more robustly at higher affinities11. Other experiments examining antitumor immune responses and autoimmunity confirmed these results and identified 10 μM affinity as the optimal affinity for interaction with a TAA12.
Many TAAs are unmutated self-antigens13,14, which include post-translationally altered-self, oncofetal embryonic, cancer testes, idiotypic/clonal, and overexpressed tissue differentiation antigens (reviewed in15). As a result of negative selection, T cells targeting TAAs express TCRs with lower affinity for their antigenic ligands as compared to pathogenic and autoimmune T cells (summarized in Figure 1)16. For example, the prototypical TAA TRP-1 is expressed in the thymus by the transcription factor AIRE17. As discussed below, these lower affinity interactions differently stimulate CD8+ T cells and have suboptimal, somewhat truncated immune responses18. Results in both human and mouse studies have proposed that high functional avidity and affinity thresholds of the TCR are required for efficient antitumor immunity19. Lower affinity interactions, like those corresponding to normal tissue antigens, can go unrecognized. To analyze the importance of T cell affinity in cancer immunotherapy, appropriate models are necessary.
Figure 1.

Average T cell receptor binding affinities to differing types of antigen-MHC complexes in the thymus and periphery. Self antigen, TAA, and positive selection are considered low-affinity TCR-pMHC interactions.
Detecting T cell receptor affinity
T cell affinity is defined as the probability that a given TCR will interact with a pMHC ligand. The strength of this interaction predicts how the T cell functions when it comes in contact with a specific antigen. Affinity is commonly reported as an equilibrium dissociation constant KD, which is the ratio of the kinetic off (koff) and on (kon) rates. The smaller the KD value, the stonger the equilibrium binding affinity of the ligand to its receptor. The interaction between the ectodomains of the TCR and pMHC is measured as the amount of free, unbound TCR in equilibrium with bound TCR-pMHC and is independent of ligand:receptor concentrations or kinetic rates3,20. The half life (t1/2) or dwell time of an interaction is a kinetic measurement derived from the rate of dissociation of the TCR-pMHC interaction. Typically the KD is proportional to the t1/2, but there are also reports of disparate values between the two3. Low affinity interactions, like many TCR-pMHC interations, are often characterized by rapid dissociation rates, which can make it difficult to use kinetic measurements to determine affinity21. One method to estimate the KD under these conditions is to use Scatchard plots. The measurements of KD, koff, kon, and t1/2 have been used to describe many different facets of TCR and T cell function (reviewed in3). Here, we do not try to cover all of this research, but focus on the KD, or binding equilibrium constant, of reported “low affinity” interactions.
One standard technique to determine these binding constants is surface plasmon resonance (SPR). SPR allows a ligand in solution to associate to an anchored receptor (or solution phase receptor to bind to an anchored ligand), resulting in a change in mass on the immobilized molecule. The affinity constants are determined by measuring the change in instrument response by varying the concentration of the ligand in solution in real time. Although SPR is valuable to make affinity measurements, T cell activation depends on numerous other factors (such as other molecules, membrane confinement, immune synapse formation) for signaling, and affinity measurements do not account for any additional forces, such as co-receptor binding22. The CD8 co-receptor modifies the measured affinity of the T cell receptor up to ten-fold23. As mentioned above, the equilibrium constant KD by definition only measures the affinity of the TCR ectodomain portion of the T cell receptor to a given ligand. However, the measured affinity between the entire TCR and pMHC is modulated in the context of the entire TCR-CD3 multi-subunit complex. Therefore, depending on the question being asked, techniques to predict relative binding in the context of the whole T cell interaction with an antigen presenting cell may be more appropriate, such as FRET, FRAP, or the 2D micropipette adhesion frequency assay (2D-MP)24,25. Lastly, in addition to SPR, affinity measurements for low-affinity interactions can be enhanced using thermodynamic techniques, such as titration calorimetry21.
In addition to direct measurements, affinity is often indirectly detected by manipulation of the TCR ligands. Such measurements of the collective strength of the functional and physical interactions leading to T cells responses are also known as “functional avidity.” Detection of an antigen-specific T cell is commonly performed using soluble recombinant multimeric pMHC. pMHC molecules are standardly multimerized into dimers, tetramers, pentamers or octamers26; typically, monomeric pMHC does not bind with high enough affinity to detect binding. Multimers are conjugated to a fluorophore for detection via flow cytometry or via a barcode for single cell analysis. Intensity of staining with multimers can act as a proxy for affinity measurements; the higher affinity T cells bind the tetramer better than lower affinity T cells, assuming similar expression of TCR and co-receptor27–29. However, higher tetramer binding does not always correlate with functional T cell responses30,31. For example, upon activation, TRP1-tetramerLO and TRP1-tetramerHI T cells similarly arrest tumor growth in a B16 model30. In addition, if the T cells being compared express different amounts of TCR or co-receptor, the affinity correlation may not hold true. Multimer detection has recently been described to introduce a bias towards high-affinity interactions, underestimating the lower affinity components25. Because these reagents are multimerized, the avidity of the interaction biases the interpretation of what the affinity of multimer-specific T cells may be. Therefore, a more accurate interpretation of multimer-binding would be detection of functional avidity, not binding affinity.
Many signaling molecules and transcription factors act as rheostats downstream of the TCR-pMHC interaction to control T cell function. These molecules translate the strength of the interaction between TCR and pMHC into a T cell response. Many of these rheostat experiments were performed using altered peptide ligands (APLs) of the SIINFEKL peptide, which is encoded by the chicken ovalbumin protein32. These peptides complex with the murine H-2Kb MHC molecule and are recognized by the CD8+ OT-1 transgenic T cell. Negative costimulatory molecules such as CD533–35 and tyrosine phosphatase PTPN2236 have been found to inhibit T cell signaling in an affinity-dependent manner using the OT-1 system. CD5 expression in the periphery appears to be linked to THEMIS, a TCR signalosome component. THEMIS is constitutively associated with the phosphatases Shp1 and Shp2 to regulate signals from very low-affinity pMHC interactions by altering cellular metabolism that drive CD8+ T cell proliferation37,38. THEMIS also plays an important role in the positive selection of low affinity T cells (reviewed in39). The phosphatase PTPN22 was found to limit conjugate formation between LFA-1 on the T cell and the ICAM ligand molecule on the antigen presenting cell; knocking out the LFA-1 gene in the T cell altered the activation threshold36. Similarly, using the OT-1 APL system, it was determined that the transcription factor IRF4 is critical for increasing glycolysis and metabolic function required for T cell activation of high-affinity T cells; low-affinity T cells show diminished glucose uptake40. Lastly, the orphan receptor Nur77 is commonly used as a marker for TCR signal strength10,41. In the OT-1 system using a Nur77GFP-reporter mouse, Nur77 expression increased as TCR-pMHC affinity increased in the thymus10. The effect of these rheostats was determined using transgenic mice specific for a foreign antigen and elucidates the usefulness of these approaches.
Current murine affinity models
Altered peptide ligands of both human and mouse native antigens are used to model TAAs. Low-affinity and/or positive selecting ligands in mice transgenic for the antigen and/or the TCRs may mimic endogenous low-affinity interactions. Much of the TCR affinity research has been performed in genetically pliable mice and has investigated the T cell response to model foreign antigens. However, the foreign transgenes used in these studies present thymic-selection-related limitations. Additionally, the binding affinities of bona fide tumor antigens are often lower than what a transgenic or altered peptide model can provide. A table of the affinities of native MHC class I-restricted peptides and their corresponding APLs for use in transgenic CD8+ T cell models is provided (Table 142–52.) This list summarizes the low-affinity altered peptides and TCRs available for use in murine in vivo oncoimmunotherapy affinity models.
Table 1.
Transgenic CD8+ T cell mouse models with known high and low affinity TCR-pMHCI interactions
| TCR | Peptide/MHC | Peptide Sequence | KD (μM) | Ref. |
|---|---|---|---|---|
| OT-1 | OVA/Kb | SIINFEKL | 6.1 | Yachi et al. |
| OT-1 | A2/Kb | SAINFEKL | 4.4 | Yachi et al. |
| OT-1 | G4/Kb | SIIGFEKL | 10.1 | Yachi et al. |
| OT-1 | E1/Kb | EIINFEKL | 20.8 | Yachi et al. |
| OT-1 | R4/Kb | SIIRFEKL | 48.9 | Yachi et al. |
| CT | A5/Ld | SPSYAYHQF | 1.2 | Slansky et al. |
| CT | AH1/Ld | SPSYVYHQF | 5 | Slansky et al. |
| ID4 | A5/Ld | SPSYAYHQF | 5 | Burhman et al. |
| ID4 | AH1/Ld | SPSYVYHQF | >100 | Burhman et al. |
| 2C | p2Ca/Ld | LSPFPFDL | 3.3 | Garcia et al. |
| 2C | QL9/Ld | QLSPFPFDL | 3.9 | Garcia et al. |
| 2C | SIY/Kb | SIYRYYGL | 31.9 | Garcia et al. |
| 2C | dEV8/Kb | EQYKFYSV | 84 | Garcia et al. |
| AHIII | p1058/Db | FAPGFFPYL | 81.4 | Buslepp et al. |
| AHIII | p1049/HLA-A2 | ALWGFFPVL | 11.3 | Buslepp et al. |
| P14 | gp33/Db | KAVYNFATC | 12.2 | Tian et al. |
| P14 | Y4A/Db | KAVANFATC | 63 | Tian et al. |
| P14 | KIMC9M/Db | KIMYNFATM | 74.5 | Tian et al. |
| P14 | KIR/Db | KIRYNFATC | 81.6 | Tian et al. |
| P14 | KISC9M/Db | KISYNFATM | 267 | Tian et al. |
| P14 | V3LC9M/Db | KALYNFATM | 264 | Tian et al. |
| P14 | Y4SC9M/Db | KAVSNFATM | 264.1 | Tian et al. |
| HY | HY/Db | KCSRNRQYL | 171 * | Maile et al. |
| HY | C2A/Db | KASRNRQYL | 221 * | Maile et al. |
| HY | K1A/Db | ACSRNRQYL | 1159 * | Maile et al. |
| TRP1 | M9/Db | TAPDNLGYM | NA - peptide stabilization | Clancy-Thompson et al. |
| TRP1 | K8/Db | TAPDNLGKM | NA - peptide stabilization | Clancy-Thompson et al. |
| PMEL | hgp100/Db | KVPRNQDWL | NA - peptide stabilization | Overwijk et al. |
| PMEL | mgp100/Db | EGSRNQDWL | NA - peptide stabilization | Overwijk et al. |
| CL4 | HA/Kd | IYSTVASSL | NA - EC50 410pM | Hartemann-Heurtier et al. |
| CL4 | A517G/Kd | IYSTVVSSL | NA - EC50 15pM | Hartemann-Heurtier et al. |
| N-TgN HI | RNEU/Dq | PDSLRDLSVF | NA – tetramer | Weiss et al. |
| N-TgNLO | RNEU/Dq | PDSLRDLSVF | NA - tetramer | Weiss et al. |
binding affinity in female mice
In addition to these reported transgenic models, there are many other systems in which affinity has been modeled. When searching for native ligands and APLs for human T cells, CD4+ T cells, or modulating affinity in a system by altering the T cell instead of the peptide, a variety of manually curated databases exist. Webservers such as ATLAS (Altered TCR Ligand Affinities and Structures) serve as an important resource for searching binding affinities between wild type or mutant TCRs and their corresponding antigens53. Additional databases such as VDJdb54, iMatrix55, PAComplex56 and MPID-T257 are sequence-structure-function based tools that allow users to interrogate TCR, peptide and MHC sequences, their structures, and cellular outcomes. Some of these programs can input the structural models and associated TCR and pMHC data to predict peptide binding for antigen discovery. Such programs are being developed to take bioinformatic predictions and translate them into “immunogenicity scores” for discovery of potent epitopes58. These transgenic models and predictive databases allow researchers to model affinity and can be advantageous when modeling novel and known TAAs.
Differences between low- and high-affinity TCR-pMHC interactions
Low-affinity T cells play an active role in immune responses25. The affinity of the signals thymocytes receive during thymic development dictate their pathway of differentiation. Using the OT-1 APL peptides, an affinity of 20–60 μM was found to induce positive selection, while peptides of stronger affinities (KD ~6 μM) stimulate negative selection59,60. Compartmentalization of the Ras/MAPK signaling pathway detects these threshold differences61. Once matured and in the periphery, T cells are much less sensitive to signals received through the TCR than their thymocyte counterparts.
Outcomes of signaling in mature T cells depend on the affinity of the pMHC interaction with the TCR. Early studies using mature OT-1 CD8+ T cells and APLs reported weak TCR interactions preferentially inducing homeostatic proliferation, suggesting differences in signaling62. In both the T1 and OT-1 APL mouse models, only agonist peptides form the interactions required to couple TCR binding and signaling63,64. Similarly, only the agonist peptide in this system induces CD3 conformational change in the CD3 subunits of the TCR65. Using the OT-1 APL system, low-affinity TCR-pMHC interactions are sufficient to activate naive cells, induce proliferation, and stimulate T cell maturation into effector and memory cells32. However, sustaining these signals is impaired in the low-affinity interactions and activation is slowed66. Interestingly, OT-1 transgenic mice with a point mutation in the TCRβ chain may also provide insights into how affinity influences T cell fate67. Although memory T cells are generated in these mice, memory responses to high-affinity interactions are deficient. Low-affinity ligands trigger more central memory CD8+ precursors, which express less T-bet and more Eomes transcription factors67. Weak interactions result in efficient NF-κB signaling that persist in immune responses. Using the OT-1 APL system, low-affinity-primed memory cells produce more IL-2 and fully differentiated memory cells that are CD45RBHI 68. Using specific T cells with low and high affinity for the NY-ESO-1 TAA peptide complexed with HLA-A2, the high-affinity interactions resulted in transient signal activation leading to poor MAPK production and low proliferation ability69. Taken together, these data suggest that low-affinity T cells have a separate, but necessary role in T cell function (summarized in Figure 2).
Figure 2.

Functional differences in CD8+ T cells between low- and high-affinity TCR-pMHC interactions. Relative to high-affinity T cells, low-affinity T cells play a distinct role in development, effector function, memory function and in the tumor microenvironment. Both are crucial contributors to the immune response.
The CD8 co-receptor modulates the strength of the TCR-pMHC interaction by binding to the alpha 3 domain of the MHC class I molecule70. Although the affinity of the TCR-pMHC interaction is unchanged by CD8, the overall stability of the complex is enhanced as the number of receptor-ligand interactions is increased. Low-affinity APLs delay CD8 co-receptor interactions, suggesting that CD8 binding contributes to low-affinity signals more than high-affinity signals23,42. Consistent with this framework, in a naturally occurring HCV model a high-affinity human TCR is more capable of CD8-independent functional responses than it’s low affinity counterpart70. These findings have practical implications: detection of low-affinity antigens using TCR multimers may depend more on CD8 binding than the high-affinity T cells.
Current dogma on CD8+ TCR affinity predicts that high-affinity tumor infiltrating lymphocytes (TILs) are the cells most important for tumor clearance. In a transgenic T cell model against HA, high-affinity T cells show increased effector function, express lower levels of inhibitory receptors, higher levels of activation markers, less pro-apoptotic gene expression, and more expression of effector molecule expression71. However, low-affinity TILs play a distinct role and are also necessary for tumor clearance. Using the OT-1 APL system with the B16 melanoma tumor model, an intermediate duration of TCR-pMHC interaction promoted release of cytotoxic vesicles and release of effector cytokines72. Peptides that provide too short or too long of an interaction altered tumor infiltration and provided little anti-tumor activity. TCR recognition of weak peptides during the priming phase can also change the effector function of antigen-specific TILs49. Using two lines of mice with TCRs that recognize the TAA TRP1 bound to H-2Db with differing affinities (TRP1HI and TRP1LO) and a panel of APLs in the B16 tumor model, it was shown that priming with the high-affinity peptide leads primarily to IFNγ production, while priming with a low-affinity peptide leads to a more cytolytic cell, although adoptive transfer of either significantly delays tumor growth49.
Using ID8 ovarian carcinoma cells expressing ovalbumin, adoptive cell therapy with high- and low-affinity T cell interactions were examined with T cells from OT-1 and OT-3 transgenic mice, respectively. T cells were transferred into mice that express a membrane-bound form of ovalbumin as a “self” antigen in kidney and pancreas. The OT-3 T cells mediate tumor regression with remarkable specificity for the tumor antigen and no evidence of autoimmunity73. The specificity of the interaction of the T cell with only the tumor cell is a key component of the low-affinity CD8+ T cell response in the tumor. The specificity, combined with the findings that low-affinity TAA-specific CD8+ T cells in the tumor are an active, cytolytic component of the immune response responsible for tumor clearance, suggests a necessary role for low-affinity TILs74.
Although high-affinity CD8+ T cells show decreased levels of inhibitory receptors in a transgenic model, high antigen levels in chronic infections also induce an exhausted phenotype75. T cells exhibiting high functional avidity show more signs of exhaustion when responding to an infection or tumors76. Additionally, IRF4 establishes and sustains T cell exhaustion77. With a previously described role for IRF4 in CD8+ T cell affinity40, higher affinity T cells may sustain an exhausted T cell phenotype. However, in several studies, exhaustion markers are upregulated in both low and high-affinity interactions in the tumor73,78. Therefore, the functionality of low- and high-affinity T cells may be abrogated in confounding ways by the tumor microenvironment. Taken together, low-affinity T cells play a distinct role in development, effector function, memory function, and in the tumor microenvironment and are crucial contributors to a fully functional immune response.
Altering the T cell activation threshold
Understanding the role of low-affinity interactions is imperative in learning how to augment responses to these interactions in the context of cancer immunotherapy. Various laboratories have reported that subtle changes in the variable domain of TCRβ can increase the affinity of the receptor using techniques such as directed molecular evolution79–82, structure based design83–85, and computational design86–88, among others. However, strategies to increase affinity of the receptor may not improve efficacy if a threshold affinity is already met12. Additional factors, such as antigen density, TCR-pMHC stabilization and CD8 co-receptor signaling is also necessary for predicticting anti-tumor therapeutic potential89. Adoptive cell therapy of high-affinity CD8+ T cells has had mixed results18,90,91. Enhancing affinities can also have off-target results; as the affinity for the tumor antigen increases, so does the TCR’s affinity for structurally similar peptides92. For example, an anti-MAGE-A3 engineered T cell receptor induced a large inflammatory response resulting in neuronal cell destruction and death93. Techniques such as TCR “fingerprinting” and mRNA engineered T cells have been developed to limit receptor-crossreactivity and off-target effects94,95. On the other hand, adoptive transfers of endogenous TCRs from TIL can often lead to immune escape. To overcome this hurdle, T cells expressing two additional T cell receptors (TETARs) have been developed and shown to have better efficacy than traditional adoptive transfers96. In this system, two TCRs to melanoma TAAs evade immune escape. Altering affinity at the TCR level is an important step in overcoming central tolerance.
“Peptide velcro” has recently been described as a strategy to increase the affinity of low-affinity ligands in the context of cancer, but still maintain the specificity that low-affinity interactions possess97. Here, through a combinatorial library approach, a secondary interaction site is engineered independently of the interface between the TCR and pMHC. This site provides a 10-fold affinity gain between TCR and pMHC, enhancing even extremely low affinities of > 100 μM. Similar to adoptive cell therapy, this requires genetic manipulation of patient T cells and infusion of the corrected or higher affinity cells back into the patient.
Peptide vaccination with either endogenous or mimetic antigen has both progressed and had challenges98. Vaccines with adjuvant and naturally occurring TAAs stimulate activation and proliferation of antigen-specific cells at a rate of <1% total circulating CD8+ T cells, much less than vaccines to foreign antigen99. However, vaccines to specific oncogenic viral antigens such as the HPV vaccine have been successful100. Mimetic peptides often induce greater immune responses than naturally occurring peptides and have had some success. However, T cells are sensitive to the structural differences; T cells primed with a mimetic peptide do not always recognize the wild type peptide101,102. An important consideration when using these mimetic peptides as vaccines is determining the cross-reactivity of the T cell repertoire responding to the native peptide relative to the mimetic103. Lastly, vaccination of low-affinity mTERT epitopes exhibited potent anti-tumor effects in a murine tumor model, while high-affinity epitopes provided no protection, suggesting that weak binding may be compensated for104.
In addition to affinity of the TCR to pMHC, high affinity between the peptide for MHC (10 nM) is often considered necessary for tumor regression105; although this is controversial as neoantigens do not demonstrate strong affinity towards MHC class I106. Weak binding of the peptide to the MHC molecule may be compensated with increased antigen, or high affinity TCRs as mentioned previously. Changing the affinity of the peptide for MHC and not the affinity of the TCR/pMHC interaction may prove to be an additional strategy; however, as mentioned above, one must consider the extent of cross-reactivity of the T cell repertoire responding to the native peptide relative to the peptides changed to improve binding to the MHC molecule101,107.
Low-dose radiation decreases the activation threshold of T cells, promoting anti-tumor immunity. Two to four Gy directly affects the T cells themselves, lowering the TCR activation threshold and skewing towards a Th1 population108. CD8+ T cells are especially sensitive to glycosylation differences because the CD8 molecule is highly glycosylated. Reduction of glycosylation through either enzyme treatment or pretreatment of T cells with neuraminidase increases receptor clustering and sensitivity, similarly decreasing the activation threshold of T cells without the need for adoptive cell therapy109,110.
Manipulation of TCR triggering can also lower the activation threshold in CD8+ T cells. Induction of TCR clustering by a magnetic field induces aggregation of nanoparticles bound to T cells111. This nanoparticle treatment provides a clinical application against B16 melanoma. Because bivalent or multivalent complexes are preferentially triggered at lower affinity interactions112, this treatment may work by lowering the activation threshold of the T cells. Similarly, inducing CD3 conformational change in the TCR-CD3 complex through the use of a monomeric fab fragment against CD3 also decreases the activation threshold113. In the OT-1 APL model, non-agonist stimulation was enhanced against peptides within a specific range of affinities. The many rational therapeutic designs currently being studied with the goal of augmenting low-affinity TCR-pMHC interactions in TILs highlights the importance of signal strength in immuno-oncology.
Conclusion
An important role for low-affinity T cells in cancer immunotherapy is coming to light. Understanding the hurdles with low-affinity T cells - central tolerance, weak binding, competition with higher affinity receptors and antigen - will delineate how to better design cancer immunotherapeutics against TAAs. Importantly, when studying low-affinity T cells, low affinity must be defined in the context of the model being used. Because of the many tools existing to measure affinity, as well as indirect markers of affinity, confounding results about the important role of low-affinity T cells exist. When recapitulating T cell affinity in the tumor, using antigen models more similar to TAAs may better translate to efficacy in the clinic. The contribution of studying well designed models of low-affinity T cells has significant repercussions in the age of cancer immunotherapy.
Acknowledgements
We apologize to those whose data on the subject we did not include, especially those who advanced the field using the OT-1 TCR and its APLs. There are many of you. MMH and JES are supported by CA226879.
Abbreviations
- 2D-MP
two dimensional micropipette adhesion frequency assay
- AIRE
autoimmune regulator
- pMHC
peptide-major histocompatibility complex
- SPR
surface plasmon resonance
- TAA
tumor associated antigen
- TCR
T cell receptor
- TETARS
T cells expressing two additional T cell receptors
- THEMIS
Thymocyte-expressed molecule involved in selection
- TIL
tumor infiltrating lymphocyte
References
- 1.Cheever MA, Allison JP, Ferris AS, et al. The prioritization of cancer antigens: a national cancer institute pilot project for the acceleration of translational research. Clin Cancer Res. 2009;15(17):5323–5337. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 2.Stone JD, Harris DT, Kranz DM. TCR affinity for p/MHC formed by tumor antigens that are self-proteins: impact on efficacy and toxicity. Curr Opin Immunol. 2015;33:16–22. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 3.Stone JD, Chervin AS, Kranz DM. T-cell receptor binding affinities and kinetics: impact on T-cell activity and specificity. Immunology. 2009;126(2):165–176. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 4.Joglekar AV, Leonard MT, Jeppson JD, et al. T cell antigen discovery via signaling and antigen-presenting bifunctional receptors. Nat Methods. 2019;16(2):191–198. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 5.Munson DJ, Egelston CA, Chiotti KE, et al. Identification of shared TCR sequences from T cells in human breast cancer using emulsion RT-PCR. Proc Natl Acad Sci U S A. 2016;113(29):8272–8277. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 6.Gee MH, Han A, Lofgren SM, et al. Antigen Identification for Orphan T Cell Receptors Expressed on Tumor-Infiltrating Lymphocytes. Cell. 2018;172(3):549–563 e516. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 7.Gnjatic S, Nishikawa H, Jungbluth AA, et al. NY-ESO-1: review of an immunogenic tumor antigen. Adv Cancer Res. 2006;95:1–30. [DOI] [PubMed] [Google Scholar]
- 8.Laan M, Peterson P. The many faces of aire in central tolerance. Front Immunol. 2013;4:326. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 9.Hogquist KA, Baldwin TA, Jameson SC. Central tolerance: learning self-control in the thymus. Nat Rev Immunol. 2005;5(10):772–782. [DOI] [PubMed] [Google Scholar]
- 10.Moran AE, Holzapfel KL, Xing Y, et al. T cell receptor signal strength in Treg and iNKT cell development demonstrated by a novel fluorescent reporter mouse. J Exp Med. 2011;208(6):1279–1289. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 11.Koehli S, Naeher D, Galati-Fournier V, Zehn D, Palmer E. Optimal T-cell receptor affinity for inducing autoimmunity. Proc Natl Acad Sci U S A. 2014;111(48):17248–17253. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 12.Zhong S, Malecek K, Johnson LA, et al. T-cell receptor affinity and avidity defines antitumor response and autoimmunity in T-cell immunotherapy. Proc Natl Acad Sci U S A. 2013;110(17):6973–6978. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 13.Novellino L, Castelli C, Parmiani G. A listing of human tumor antigens recognized by T cells: March 2004 update. Cancer Immunol Immunother. 2005;54(3):187–207. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 14.Coulie PG, Van den Eynde BJ, van der Bruggen P, Boon T. Tumour antigens recognized by T lymphocytes: at the core of cancer immunotherapy. Nat Rev Cancer. 2014;14(2):135–146. [DOI] [PubMed] [Google Scholar]
- 15.Blankenstein T, Coulie PG, Gilboa E, Jaffee EM. The determinants of tumour immunogenicity. Nat Rev Cancer. 2012;12(4):307–313. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 16.Dunn GP, Old LJ, Schreiber RD. The three Es of cancer immunoediting. Annu Rev Immunol. 2004;22:329–360. [DOI] [PubMed] [Google Scholar]
- 17.Su MA, Anderson MS. Breaking through the central tolerance ceiling to unleash anticancer immune responses. Oncoimmunology. 2014;3(8):e950169. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 18.Corse E, Gottschalk RA, Allison JP. Strength of TCR-peptide/MHC interactions and in vivo T cell responses. J Immunol. 2011;186(9):5039–5045. [DOI] [PubMed] [Google Scholar]
- 19.Schmid DA, Irving MB, Posevitz V, et al. Evidence for a TCR affinity threshold delimiting maximal CD8 T cell function. J Immunol. 2010;184(9):4936–4946. [DOI] [PubMed] [Google Scholar]
- 20.Zhu C, Jiang N, Huang J, Zarnitsyna VI, Evavold BD. Insights from in situ analysis of TCR-pMHC recognition: response of an interaction network. Immunol Rev. 2013;251(1):49–64. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 21.Piepenbrink KH, Gloor BE, Armstrong KM, Baker BM. Methods for quantifying T cell receptor binding affinities and thermodynamics. Methods Enzymol. 2009;466:359–381. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 22.Kuball J, Hauptrock B, Malina V, et al. Increasing functional avidity of TCR-redirected T cells by removing defined N-glycosylation sites in the TCR constant domain. J Exp Med. 2009;206(2):463–475. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 23.Liu B, Zhong S, Malecek K, et al. 2D TCR-pMHC-CD8 kinetics determines T-cell responses in a self-antigen-specific TCR system. Eur J Immunol. 2014;44(1):239–250. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 24.Hebeisen M, Allard M, Gannon PO, Schmidt J, Speiser DE, Rufer N. Identifying Individual T Cell Receptors of Optimal Avidity for Tumor Antigens. Front Immunol. 2015;6:582. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 25.Martinez RJ, Evavold BD. Lower Affinity T Cells are Critical Components and Active Participants of the Immune Response. Front Immunol. 2015;6:468. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 26.Bakker AH, Schumacher TN. MHC multimer technology: current status and future prospects. Curr Opin Immunol. 2005;17(4):428–433. [DOI] [PubMed] [Google Scholar]
- 27.Busch DH, Pamer EG. T cell affinity maturation by selective expansion during infection. J Exp Med. 1999;189(4):701–710. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 28.Crawford F, Kozono H, White J, Marrack P, Kappler J. Detection of antigen-specific T cells with multivalent soluble class II MHC covalent peptide complexes. Immunity. 1998;8(6):675–682. [DOI] [PubMed] [Google Scholar]
- 29.Yee C, Savage PA, Lee PP, Davis MM, Greenberg PD. Isolation of high avidity melanoma-reactive CTL from heterogeneous populations using peptide-MHC tetramers. J Immunol. 1999;162(4):2227–2234. [PubMed] [Google Scholar]
- 30.Dougan SK, Dougan M, Kim J, et al. Transnuclear TRP1-specific CD8 T cells with high or low affinity TCRs show equivalent antitumor activity. Cancer Immunol Res. 2013;1(2):99–111. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 31.al-Ramadi BK, Jelonek MT, Boyd LF, Margulies DH, Bothwell AL. Lack of strict correlation of functional sensitization with the apparent affinity of MHC/peptide complexes for the TCR. J Immunol. 1995;155(2):662–673. [PubMed] [Google Scholar]
- 32.Zehn D, Lee SY, Bevan MJ. Complete but curtailed T-cell response to very low-affinity antigen. Nature. 2009;458(7235):211–214. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 33.Fulton RB, Hamilton SE, Xing Y, et al. The TCR’s sensitivity to self peptide-MHC dictates the ability of naive CD8(+) T cells to respond to foreign antigens. Nat Immunol. 2015;16(1):107–117. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 34.Azzam HS, Grinberg A, Lui K, Shen H, Shores EW, Love PE. CD5 expression is developmentally regulated by T cell receptor (TCR) signals and TCR avidity. J Exp Med. 1998;188(12):2301–2311. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 35.Tarakhovsky A, Kanner SB, Hombach J, et al. A role for CD5 in TCR-mediated signal transduction and thymocyte selection. Science. 1995;269(5223):535–537. [DOI] [PubMed] [Google Scholar]
- 36.Salmond RJ, Brownlie RJ, Morrison VL, Zamoyska R. The tyrosine phosphatase PTPN22 discriminates weak self peptides from strong agonist TCR signals. Nat Immunol. 2014;15(9):875–883. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 37.Paster W, Bruger AM, Katsch K, et al. A THEMIS:SHP1 complex promotes T-cell survival. EMBO J. 2015;34(3):393–409. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 38.Brzostek J, Gautam N, Zhao X, et al. T cell receptor and cytokine signal integration in CD8(+) T cells is mediated by the protein Themis. Nat Immunol. 2020;21(2):186–198. [DOI] [PubMed] [Google Scholar]
- 39.Lyu J, Wang L, Lu L. Thymocyte selection: From signaling to epigenetic regulation. Adv Immunol. 2019;144:1–22. [DOI] [PubMed] [Google Scholar]
- 40.Man K, Miasari M, Shi W, et al. The transcription factor IRF4 is essential for TCR affinity-mediated metabolic programming and clonal expansion of T cells. Nat Immunol. 2013;14(11):1155–1165. [DOI] [PubMed] [Google Scholar]
- 41.Au-Yeung BB, Zikherman J, Mueller JL, et al. A sharp T-cell antigen receptor signaling threshold for T-cell proliferation. Proc Natl Acad Sci U S A. 2014;111(35):E3679–3688. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 42.Yachi PP, Ampudia J, Zal T, Gascoigne NR. Altered peptide ligands induce delayed CD8-T cell receptor interaction--a role for CD8 in distinguishing antigen quality. Immunity. 2006;25(2):203–211. [DOI] [PubMed] [Google Scholar]
- 43.Slansky JE, Rattis FM, Boyd LF, et al. Enhanced antigen-specific antitumor immunity with altered peptide ligands that stabilize the MHC-peptide-TCR complex. Immunity. 2000;13(4):529–538. [DOI] [PubMed] [Google Scholar]
- 44.Buhrman JD, Jordan KR, Munson DJ, Moore BL, Kappler JW, Slansky JE. Improving antigenic peptide vaccines for cancer immunotherapy using a dominant tumor-specific T cell receptor. J Biol Chem. 2013;288(46):33213–33225. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 45.Garcia KC, Tallquist MD, Pease LR, et al. Alphabeta T cell receptor interactions with syngeneic and allogeneic ligands: affinity measurements and crystallization. Proc Natl Acad Sci U S A. 1997;94(25):13838–13843. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 46.Buslepp J, Kerry SE, Loftus D, Frelinger JA, Appella E, Collins EJ. High affinity xenoreactive TCR:MHC interaction recruits CD8 in absence of binding to MHC. J Immunol. 2003;170(1):373–383. [DOI] [PubMed] [Google Scholar]
- 47.Tian S, Maile R, Collins EJ, Frelinger JA. CD8+ T cell activation is governed by TCR-peptide/MHC affinity, not dissociation rate. J Immunol. 2007;179(5):2952–2960. [DOI] [PubMed] [Google Scholar]
- 48.Maile R, Siler CA, Kerry SE, Midkiff KE, Collins EJ, Frelinger JA. Peripheral “CD8 tuning” dynamically modulates the size and responsiveness of an antigen-specific T cell pool in vivo. J Immunol. 2005;174(2):619–627. [DOI] [PubMed] [Google Scholar]
- 49.Clancy-Thompson E, Devlin CA, Tyler PM, et al. Altered Binding of Tumor Antigenic Peptides to MHC Class I Affects CD8(+) T Cell-Effector Responses. Cancer Immunol Res. 2018;6(12):1524–1536. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 50.Overwijk WW, Tsung A, Irvine KR, et al. gp100/pmel 17 is a murine tumor rejection antigen: induction of “self”-reactive, tumoricidal T cells using high-affinity, altered peptide ligand. J Exp Med. 1998;188(2):277–286. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 51.Hartemann-Heurtier A, Mars LT, Bercovici N, et al. An altered self-peptide with superagonist activity blocks a CD8-mediated mouse model of type 1 diabetes. J Immunol. 2004;172(2):915–922. [DOI] [PubMed] [Google Scholar]
- 52.Weiss VL, Lee TH, Song H, et al. Trafficking of high avidity HER-2/neu-specific T cells into HER-2/neu-expressing tumors after depletion of effector/memory-like regulatory T cells. PLoS One. 2012;7(2):e31962. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 53.Borrman T, Cimons J, Cosiano M, et al. ATLAS: A database linking binding affinities with structures for wild-type and mutant TCR-pMHC complexes. Proteins. 2017;85(5):908–916. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 54.Shugay M, Bagaev DV, Zvyagin IV, et al. VDJdb: a curated database of T-cell receptor sequences with known antigen specificity. Nucleic Acids Res. 2018;46(D1):D419–D427. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 55.Liu IH, Lo YS, Yang JM. Genome-wide structural modelling of TCR-pMHC interactions. BMC Genomics. 2013;14 Suppl 5:S5. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 56.Liu IH, Lo YS, Yang JM. PAComplex: a web server to infer peptide antigen families and binding models from TCR-pMHC complexes. Nucleic Acids Res. 2011;39(Web Server issue):W254–260. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 57.Khan JM, Cheruku HR, Tong JC, Ranganathan S. MPID-T2: a database for sequence-structure-function analyses of pMHC and TR/pMHC structures. Bioinformatics. 2011;27(8):1192–1193. [DOI] [PubMed] [Google Scholar]
- 58.Ogishi M, Yotsuyanagi H. Quantitative Prediction of the Landscape of T Cell Epitope Immunogenicity in Sequence Space. Front Immunol. 2019;10:827. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 59.Alam SM, Travers PJ, Wung JL, et al. T-cell-receptor affinity and thymocyte positive selection. Nature. 1996;381(6583):616–620. [DOI] [PubMed] [Google Scholar]
- 60.Naeher D, Daniels MA, Hausmann B, Guillaume P, Luescher I, Palmer E. A constant affinity threshold for T cell tolerance. J Exp Med. 2007;204(11):2553–2559. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 61.Daniels MA, Teixeiro E, Gill J, et al. Thymic selection threshold defined by compartmentalization of Ras/MAPK signalling. Nature. 2006;444(7120):724–729. [DOI] [PubMed] [Google Scholar]
- 62.Goldrath AW, Bevan MJ. Low-affinity ligands for the TCR drive proliferation of mature CD8+ T cells in lymphopenic hosts. Immunity. 1999;11(2):183–190. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 63.Sibener LV, Fernandes RA, Kolawole EM, et al. Isolation of a Structural Mechanism for Uncoupling T Cell Receptor Signaling from Peptide-MHC Binding. Cell. 2018;174(3):672–687 e627. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 64.Liu B, Chen W, Evavold BD, Zhu C. Accumulation of dynamic catch bonds between TCR and agonist peptide-MHC triggers T cell signaling. Cell. 2014;157(2):357–368. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 65.Gil D, Schrum AG, Daniels MA, Palmer E. A role for CD8 in the developmental tuning of antigen recognition and CD3 conformational change. J Immunol. 2008;180(6):3900–3909. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 66.Zehn D, Bevan MJ. T cells with low avidity for a tissue-restricted antigen routinely evade central and peripheral tolerance and cause autoimmunity. Immunity. 2006;25(2):261–270. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 67.Knudson KM, Hamilton SE, Daniels MA, Jameson SC, Teixeiro E. Cutting edge: The signals for the generation of T cell memory are qualitatively different depending on TCR ligand strength. J Immunol. 2013;191(12):5797–5801. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 68.Krummey SM, Martinez RJ, Andargachew R, et al. Low-Affinity Memory CD8+ T Cells Mediate Robust Heterologous Immunity. J Immunol. 2016;196(6):2838–2846. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 69.Presotto D, Erdes E, Duong MN, et al. Fine-Tuning of Optimal TCR Signaling in Tumor-Redirected CD8 T Cells by Distinct TCR Affinity-Mediated Mechanisms. Front Immunol. 2017;8:1564. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 70.Williams CM, Schonnesen AA, Zhang SQ, et al. Normalized Synergy Predicts That CD8 Co-Receptor Contribution to T Cell Receptor (TCR) and pMHC Binding Decreases As TCR Affinity Increases in Human Viral-Specific T Cells. Front Immunol. 2017;8:894. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 71.Bos R, Marquardt KL, Cheung J, Sherman LA. Functional differences between low- and high-affinity CD8(+) T cells in the tumor environment. Oncoimmunology. 2012;1(8):1239–1247. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 72.Riquelme E, Carreno LJ, Gonzalez PA, Kalergis AM. The duration of TCR/pMHC interactions regulates CTL effector function and tumor-killing capacity. Eur J Immunol. 2009;39(8):2259–2269. [DOI] [PubMed] [Google Scholar]
- 73.Miller AM, Bahmanof M, Zehn D, Cohen EE, Schoenberger SP. Leveraging TCR Affinity in Adoptive Immunotherapy against Shared Tumor/Self-Antigens. Cancer Immunology Research. 2019;7(1):40–49. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 74.Ostroumov D, Fekete-Drimusz N, Saborowski M, Kuhnel F, Woller N. CD4 and CD8 T lymphocyte interplay in controlling tumor growth. Cell Mol Life Sci. 2018;75(4):689–713. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 75.Utzschneider DT, Alfei F, Roelli P, et al. High antigen levels induce an exhausted phenotype in a chronic infection without impairing T cell expansion and survival. J Exp Med. 2016;213(9):1819–1834. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 76.Vigano S, Utzschneider DT, Perreau M, Pantaleo G, Zehn D, Harari A. Functional avidity: a measure to predict the efficacy of effector T cells? Clin Dev Immunol. 2012;2012:153863. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 77.Man K, Gabriel SS, Liao Y, et al. Transcription Factor IRF4 Promotes CD8(+) T Cell Exhaustion and Limits the Development of Memory-like T Cells during Chronic Infection. Immunity. 2017;47(6):1129–1141 e1125. [DOI] [PubMed] [Google Scholar]
- 78.Shakiba M, Philip M, Camara S, Socci N, Schietinger A. The impact of TCR affinity on T cell differentiation and dysfunction in tumors. J Immunol. 2018;200(57.24). [Google Scholar]
- 79.Sharma P, Kranz DM. Subtle changes at the variable domain interface of the T-cell receptor can strongly increase affinity. J Biol Chem. 2018;293(5):1820–1834. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 80.Liddy N, Bossi G, Adams KJ, et al. Monoclonal TCR-redirected tumor cell killing. Nat Med. 2012;18(6):980–987. [DOI] [PubMed] [Google Scholar]
- 81.Chlewicki LK, Holler PD, Monti BC, Clutter MR, Kranz DM. High-affinity, peptide-specific T cell receptors can be generated by mutations in CDR1, CDR2 or CDR3. J Mol Biol. 2005;346(1):223–239. [DOI] [PubMed] [Google Scholar]
- 82.Schmitt TM, Aggen DH, Ishida-Tsubota K, Ochsenreither S, Kranz DM, Greenberg PD. Generation of higher affinity T cell receptors by antigen-driven differentiation of progenitor T cells in vitro. Nat Biotechnol. 2017;35(12):1188–1195. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 83.Hellman LM, Foley KC, Singh NK, et al. Improving T Cell Receptor On-Target Specificity via Structure-Guided Design. Mol Ther. 2019;27(2):300–313. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 84.Malecek K, Grigoryan A, Zhong S, et al. Specific increase in potency via structure-based design of a TCR. J Immunol. 2014;193(5):2587–2599. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 85.Pierce BG, Haidar JN, Yu Y, Weng Z. Combinations of affinity-enhancing mutations in a T cell receptor reveal highly nonadditive effects within and between complementarity determining regions and chains. Biochemistry. 2010;49(33):7050–7059. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 86.Pierce BG, Hellman LM, Hossain M, et al. Computational design of the affinity and specificity of a therapeutic T cell receptor. PLoS Comput Biol. 2014;10(2):e1003478. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 87.Mosch A, Raffegerst S, Weis M, Schendel DJ, Frishman D. Machine Learning for Cancer Immunotherapies Based on Epitope Recognition by T Cell Receptors. Front Genet. 2019;10:1141. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 88.Harris DT, Wang N, Riley TP, et al. Deep Mutational Scans as a Guide to Engineering High Affinity T Cell Receptor Interactions with Peptide-bound Major Histocompatibility Complex. J Biol Chem. 2016;291(47):24566–24578. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 89.Spear TT, Wang Y, Foley KC, et al. Critical biological parameters modulate affinity as a determinant of function in T-cell receptor gene-modified T-cells. Cancer Immunol Immunother. 2017;66(11):1411–1424. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 90.McMahan RH, McWilliams JA, Jordan KR, Dow SW, Wilson DB, Slansky JE. Relating TCR-peptide-MHC affinity to immunogenicity for the design of tumor vaccines. J Clin Invest. 2006;116(9):2543–2551. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 91.Corse E, Gottschalk RA, Krogsgaard M, Allison JP. Attenuated T cell responses to a high-potency ligand in vivo. PLoS Biol. 2010;8(9). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 92.Holler PD, Chlewicki LK, Kranz DM. TCRs with high affinity for foreign pMHC show self-reactivity. Nat Immunol. 2003;4(1):55–62. [DOI] [PubMed] [Google Scholar]
- 93.Morgan RA, Chinnasamy N, Abate-Daga D, et al. Cancer regression and neurological toxicity following anti-MAGE-A3 TCR gene therapy. J Immunother. 2013;36(2):133–151. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 94.Bentzen AK, Such L, Jensen KK, et al. T cell receptor fingerprinting enables in-depth characterization of the interactions governing recognition of peptide-MHC complexes. Nat Biotechnol. 2018. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 95.Mensali N, Myhre MR, Dillard P, et al. Preclinical assessment of transiently TCR redirected T cells for solid tumour immunotherapy. Cancer Immunol Immunother. 2019;68(8):1235–1243. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 96.Hofflin S, Prommersberger S, Uslu U, et al. Generation of CD8(+) T cells expressing two additional T-cell receptors (TETARs) for personalised melanoma therapy. Cancer Biol Ther. 2015;16(9):1323–1331. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 97.Gee MH, Sibener LV, Birnbaum ME, et al. Stress-testing the relationship between T cell receptor/peptide-MHC affinity and cross-reactivity using peptide velcro. Proc Natl Acad Sci U S A. 2018;115(31):E7369–E7378. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 98.Li W, Joshi MD, Singhania S, Ramsey KH, Murthy AK. Peptide Vaccine: Progress and Challenges. Vaccines (Basel). 2014;2(3):515–536. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 99.Hollingsworth RE, Jansen K. Turning the corner on therapeutic cancer vaccines. NPJ Vaccines. 2019;4:7. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 100.Paavonen J, Naud P, Salmeron J, et al. Efficacy of human papillomavirus (HPV)-16/18 AS04-adjuvanted vaccine against cervical infection and precancer caused by oncogenic HPV types (PATRICIA): final analysis of a double-blind, randomised study in young women. Lancet. 2009;374(9686):301–314. [DOI] [PubMed] [Google Scholar]
- 101.Madura F, Rizkallah PJ, Holland CJ, et al. Structural basis for ineffective T-cell responses to MHC anchor residue-improved “heteroclitic” peptides. Eur J Immunol. 2015;45(2):584–591. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 102.Dyson J T-cell receptors: tugging on the anchor for a tighter hold on the tumor-associated peptide. Eur J Immunol. 2015;45(2):380–382. [DOI] [PubMed] [Google Scholar]
- 103.Jordan KR, McMahan RH, Kemmler CB, Kappler JW, Slansky JE. Peptide vaccines prevent tumor growth by activating T cells that respond to native tumor antigens. Proc Natl Acad Sci U S A. 2010;107(10):4652–4657. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 104.Gross DA, Graff-Dubois S, Opolon P, et al. High vaccination efficiency of low-affinity epitopes in antitumor immunotherapy. J Clin Invest. 2004;113(3):425–433. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 105.Engels B, Engelhard VH, Sidney J, et al. Relapse or eradication of cancer is predicted by peptide-major histocompatibility complex affinity. Cancer Cell. 2013;23(4):516–526. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 106.Brennick CA, George MM, Srivastava PK, Karandikar SH. Prediction of cancer neoepitopes needs new rules. Semin Immunol. 2020:101387. [DOI] [PubMed] [Google Scholar]
- 107.Borbulevych OY, Baxter TK, Yu Z, Restifo NP, Baker BM. Increased immunogenicity of an anchor-modified tumor-associated antigen is due to the enhanced stability of the peptide/MHC complex: implications for vaccine design. J Immunol. 2005;174(8):4812–4820. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 108.Spary LK, Al-Taei S, Salimu J, et al. Enhancement of T cell responses as a result of synergy between lower doses of radiation and T cell stimulation. J Immunol. 2014;192(7):3101–3110. [DOI] [PubMed] [Google Scholar]
- 109.Starr TK, Daniels MA, Lucido MM, Jameson SC, Hogquist KA. Thymocyte sensitivity and supramolecular activation cluster formation are developmentally regulated: a partial role for sialylation. J Immunol. 2003;171(9):4512–4520. [DOI] [PubMed] [Google Scholar]
- 110.Haxho F, Neufeld RJ, Szewczuk MR. Neuraminidase-1: a novel therapeutic target in multistage tumorigenesis. Oncotarget. 2016;7(26):40860–40881. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 111.Perica K, Tu A, Richter A, Bieler JG, Edidin M, Schneck JP. Magnetic field-induced T cell receptor clustering by nanoparticles enhances T cell activation and stimulates antitumor activity. ACS Nano. 2014;8(3):2252–2260. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 112.Schamel WW, Arechaga I, Risueno RM, et al. Coexistence of multivalent and monovalent TCRs explains high sensitivity and wide range of response. J Exp Med. 2005;202(4):493–503. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 113.Hoffmann MM, Molina-Mendiola C, Nelson AD, et al. Co-potentiation of antigen recognition: A mechanism to boost weak T cell responses and provide immunotherapy in vivo. Sci Adv. 2015;1(9):e1500415. [DOI] [PMC free article] [PubMed] [Google Scholar]
