Summary
Human T cells have a diverse T cell receptor (TCR) repertoire that endows them with the ability to identify and defend against a broad spectrum of antigens. The universe of possible antigens that T cells may encounter, however, is even larger. To effectively surveil such a vast universe, the T cell repertoire must adopt a high degree of cross-reactivity. Likewise, antigen-specific and cross-reactive T cell responses play pivotal roles in both protective and pathological immune responses in numerous diseases. In this review, we explore the implications of these antigen-driven T cell responses, with a particular focus on CD8+ T cells, using infection, neurodegeneration, and cancer as examples. We also summarize recent technological advances that facilitate high-throughput profiling of antigen-specific and cross-reactive T cell responses experimentally, as well as computational biology approaches that predict these interactions.
Keywords: Antigen-specific T cells, T cell cross-reactivity, Cancer, Infection, Vaccination, Neurodegenerative diseases
TCR repertoire size, T cell number, and antigen space
The discovery of TCR genes almost 40 years ago marks the beginning of an exciting era in immunology1,2. Using V(D)J recombination, human T cells generate an enormously large repertoire of T cell antigen receptors (TCR). The estimated potential diversity could be in the range of 2×1019 unique receptors,3,4 orders of magnitude larger than the TCR repertoire of any individual. Jenkin et al5, did an exhaustive count of naïve T cells in mice, estimating that an adult mouse has about 8×107 α/β TCR+ T cells in the secondary lymphoid organs and another 5×106 in the blood. Of these, about 70% of the cells are of a naïve phenotype 5. Thus, about 2.6×107 non-naïve T cells exist in secondary lymphoid organs and blood of healthy mice under specific pathogen-free conditions. Tissue resident memory T cells constitute a major population in both mice and humans6,7. Using a quantitative immunofluorescence microscopy8 strategy to analyze the distribution of CD8 T cells in a TCR (P14) transgenic mouse 120–150 days after LCMV infection, it was estimated that about 43% of the memory P14 pool was localized in secondary lymphoid organs, 29% in circulating blood and marginated pool (including i.v. Ab+ cells from all tissues examined), and 28% in nonlymphoid tissues (including i.v. Ab− cells within liver, lung, kidney, pancreas, salivary gland, uterus, vagina and cervix, small intestine, large intestine, stomach, and thymus). Combining these two pieces of information and taking 2:1 for CD4 to CD8 T cell ratio, there are about 0.9×107 T cells in nonlymphoid tissues. This puts the total number of T cells in a mouse to be around 9.4×107. By extrapolation based on body weight, an adult human has about 4.7×1011 T cells, which is in the same order of magnitude estimated by human T cell weight9,10.
Don Mason showed an elegant derivation of the size of potential antigen space by considering the fraction of possible peptide binding to MHC11. But even a conservative estimate of 12 amino acids in peptide length and 20 possible amino acids in each position puts a rough estimate of the potential antigen space to 4.1×1015. This is about four orders of magnitude larger than the total number of T cells in a human. Thus, in order to avoid holes in TCR coverage, an individual T cell must recognize at least 10,000 antigens. The degree of cross-reactivity is likely to increase as the T cell clonal expansion significantly reduces the number of TCR diversity in the total number of T cells each individual harbors12–18. Another complexity is that single antigens are recognized by polyclonal T cells. It is extremely inefficient for each antigen to be recognized by only one cell as studies in mice and humans in both CD4 and CD8 T cell compartments12–18 have shown that precursor T cell frequency is about 1 in 105 to 1 in 106.
As a result, the interactions between a TCR repertoire and the antigen space can be described as a complex and dynamic mesh network, where multiple TCRs can recognize a single antigen and multiple antigens can be recognized by a single TCR. The interactions between TCRs and antigens in a given context greatly impact how T cells surveil, contribute to disease protection, or become dysregulated in disease. Exploring examples from TCR-antigen interaction across infection, neurodegeneration, and cancer highlights the complexity and importance of understanding the context-dependent antigen specific and cross-reactive T cell response. Therefore, this review will cover data published across these fields to explore: (1) Methodologies to identify antigen-specific T cells and their fundamental properties such as TCR affinity and functionality; (2) Examples of variable antigen-specific and cross-reactive T cell responses in infection, neurodegeneration, and cancer; (3) Computational approaches in TCR-antigen binding prediction; (4) An outlook of experimental and computational needs to facilitate TCR-antigen discovery.
Technologies in peptide antigen and antigen-specific TCR discovery
Since the paradigm-shifting finding that TCR recognizes peptides presented by MHCs19–26, there has been a widespread interest to identify peptide antigens and their cognate T cells. This effort has been facilitated by the development of mass spectrometry27. Mass-spectrometry-based immunopeptidomics has also enabled the discovery of noncanonical antigens28—antigens derived from sequences outside protein-coding regions, such as cryptic peptide discovered in neurodegenerative diseases and cancer (see below), or generated by noncanonical antigen-processing mechanisms, such as some of the cancer antigens (see below) or fusion peptides, such as those discovered in type 1 diabetes29. Recently, tremendous advances have been made in resolving proteins in a small number of cells. Multiple improvements have been made to increase the sensitivity and throughput of single cell proteome measurement30. At the same time, various technologies have been developed for single-molecule protein sequencing31. These technological advancements will undoubtedly change the landscape of antigen discovery. On the other side, many approaches to exchange peptides bound to MHC molecules have been developed32–36 as well as higher valency of peptide-MHC (pMHC) multimers37,38 and affinity matured MHC-II molecules39,40 to enhance the binding to lower affinity TCRs, especially on CD4+ T cells. Combining technologies from these areas will open many possibilities of high-throughput antigen-specific and cross-reactive TCR discovery in future studies.
There are two main categories of methods for antigen-specific TCR discoveries: pMHC multimer binding based and functional test based. In the last decade, significant progress was made in both categories and was comprehensively summarized in a recent review by Joglekar et al41. The application of some of these methods is discussed throughout the current review. Recently, there are additional technologies developed in pairing TCR with antigens. Dahotre et al42 developed a droplet digital PCR based method to count DNA barcoded pMHC bound on T cells, which could be more accurate in numerating antigen specific T cells than sequencing-based methods. Ma et al43 combined DNA-barcoded pMHC tetramer linked TCR sequencing with single cell gene expression and DNA-barcoded phenotyping antibodies to develop a multi-dimensional integrated profiling of antigen-specific T cells, named TetTCR-SeqHD. TetTCR-SeqHD enables the direct profiling of phenotypes and functional states of antigen specific T cells without any stimulation, which is critical in studying the roles of antigen specific T cells to disease initiation and pathogenesis. Liu et al44 developed a FucoID method that uses glycosyltransferase-mediated tagging to label and capture antigen-specific T cells in a cell interaction-dependent manner. Using an updated in vitro T cell expansion method, Arnaud et al45 developed NeoScreen to increase the efficiency of in vitro expansion of cancer antigen specific T cells infiltrated to tumor. This, in combination with the above technologies mentioned, could significantly increase cancer antigen specific TCR discovery. V-CARMA46 and ENTER47 represent another group of methods recently developed that take advantage of lentiviral-based display and delivery platform to identify and isolate antigen-specific T cells, and deliver cell-specific genetic cargo at the same time. Although the throughput of generating pMHC expressing viral particles is still limited, V-CARMA and ENTER are great methods to deliver cargo to T cells in an antigen-specific manner.
In addition to chemical interactions, mechanical interactions among macromolecules have emerged as another modality that impacts receptor-ligand interactions. A classic example can be found within adhesion molecules, such as selectins and integrins, that need to overcome sheer stress when helping leukocytes exit blood vessels. Initial increase of force results in an increase of bond lifetime between adhesion molecules and their ligands (catch phase). However, further increase of force results in a decrease of bond lifetime between adhesion molecules and their ligand (slip phase). Thus, the molecular interactions that contain both catch phase and slip phase are known as “catch bonds”, while the molecular interactions that contain only the slip phase are known as “slip bonds”. Slip bonds48 and catch bonds49 were predicted theoretically in 1978 and 1988, respectively. Although slip bonds were detected in many molecules, the catch bond was only first detected by Cheng Zhu’s lab in 2003 in selectins50 then again in integrins51 when bond lifetime measurements were developed and implemented. Later, it was demonstrated that TCRs also exhibit a catch-bond property when interacting with agonist and partial agonist ligands, but exhibit slip bonds when interacting with antagonist and weak agonist ligands52. Recently, it was shown that catch bonds could be used to distinguish specific antigens from cross-reactive antigens53 (see below). Thus, catch bond analysis could be integrated into TCR based therapeutic development.
Large scale of epitope screens on pathogens
Understanding the epitopes that T cells target during infection and vaccination is of great value to understanding the T cell responses and designing better vaccines. Using a variety of approaches, including multiplexing pMHC multimers with flow cytometry54, mass cytometry55–57 or NGS58 as readouts, immunopeptidomics59, activation-induction60–63, antigen presentation array-based64 assays, and others reviewed recently41, a litany of T cell targets to a variety of pathogens have been unveiled. These types of screens have laid the groundwork to deepen our understanding of the principles that govern TCR:pMHC interactions, the phenotypes that ensue, and how to intervene in order to induce more favorable outcomes.
SARS-2 and hCOVs/pre-existing immunity
One perplexing aspect of COVID-19 is the wide variability in disease severity. An early hypothesis was that memory specific to other coronaviruses that cause the common cold (hCOVs) could cross-react to SARS-CoV-2 and provide some protection. Early investigation of infections in healthcare workers revealed that individuals with abortive seronegative SARS-CoV-2 titers exhibited an expansion of pre-existing SARS-CoV-2 specific T cells in the blood65. A later study built on this rationale, finding T cells specific to the same antigens in the airways of pre-pandemic samples that correlated to the response to corresponding hCOV antigens66.
Several other studies have also corroborated the existence of pre-existing T cell immunity in the blood. Using both pMHC tetramer and stimulation-based assays, several groups have observed non-naïve, spike and non-spike specific T cells in the blood of pre-pandemic individuals over several HLA backgrounds67–69. These SARS-CoV-2 specific cells were often of an effector memory phenotype and could exhibit T cell effector functions, suggesting a potential capacity to play a role during infection. In one of these studies, Schulian, et al also showed that these cross-reactive TCRs were of comparable affinity to SARS-CoV-2 and hCOV specific TCRs. Using flow cytometry70 and single cell sequencing-based tetramer approaches58, two independent studies revealed that the degree of pre-existing cross-reactivity was dependent on the HLA background. Both studies implicate a nucleocapsid protein-derived, HLA-B*07:02-restricted (B7/NP105) epitope as highly dominant, eliciting expanded T cells in both pre-existing and post-infection repertoires (Table 1). Interestingly, both studies report a limited number of shared TCR features across donors compared to other SARS-CoV-2 epitopes, underpinned by distinct V/J gene usage and promiscuous α/β chain pairing. Nguyen et al70 also observed differences in the number of N-insertions in the CDR3s of pre-pandemic versus post-infection TCRs specific to B7/NP105, suggesting differential pressures on the repertoires before and after COVID-19.
Table 1:
Select cross-reactivity examples. Epitopes derived from various antigens and their alignments are represented58,61,86,89–91.
Epitope | Source | Sequences |
---|---|---|
B7/NP105 | SARS-CoV-2 HKU1, 0C43 (hCOVs) |
SPRWYFYYL I I I I I I I I LPRWYFYYL |
A24/S448 | WT (SARS-CoV-2) DELTA, BA.4/5 |
NYNYLYRLF I I I I I I I I NYNYRYRLF |
B37/NP338 | H3N2 H1N1 pH1N1, H5N2. H7N9 |
FEDLELLSF I I I I I I I I FEDLRVLSF I I I I I I I I FEDLRVSSF |
B35/pp65123 B35/S1095 |
CMV SARS-CoV-2 |
IPSINVHHY I I FVSNGTHWF |
NF-M18–30 MOG35–55 NF-M225–237 |
Mouse (self) | TETRSSFSRVSGS I I I I I I MEVGWYRSPFSRVVHLYRNG I I I I LQDEVAFLRSNHE |
Given that B7/NP105 is only a single amino acid point mutation from other beta coronaviruses, it is possible that differences in the antigen presentation capacity of different HLA backgrounds may dictate the degree of pre-existing immunity, thereby impacting disease severity. In fact, another large study found an association between mild disease and a B7/NP105 response71. In addition to observing usage of pre-existing immunity, high functional avidity, and effector functions, the authors also reported stronger maintenance of memory in convalescence compared to other epitopes. Possibly related to the abortive seronegative healthcare workers described early in the pandemic65, Kendu et al reported a larger number of IL2-secreting nucleocapsid specific T cells in individuals before and after close contact COVID-19 exposures who remained PCR-negative versus those who tested positive72. Although the authors did not test a direct relationship with HLA background, it is possible that some of the protection provided in their study was due to a strong response by pre-existing B7/NP105 specific T cells. It is thus possible that prior stimulation through prior encounters with hCOVs causes B7/NP105 specific cells to engage in a more robust secondary immune response and long-lived phenotypic state. Furthermore, Nguyen et al also showed that B7/NP105 specific T cells can respond to the variants of concern (VOCs) at the time (Alpha, Beta, Gamma, Delta), suggesting a capacity to continue providing protection under the selective pressure of a mutating virus70.
Cross-reactivity between SARS-CoV-2 and persistent viruses
Several risk factors have emerged as predictors of developing severe COVID-19. HIV infection73, cancer74, Type 1 and 2 Diabetes75, age76, and obesity77 are some of many pre-dispositions associated with cases of severe COVID-19. One less clear, yet interesting, example of COVID-19 comorbidity is infection with persistent viruses. Persistent viral infections, such as Epstein-Barr virus (EBV) and cytomegalovirus (CMV), are common in the population. In health they generally pose no threat, but in the immunocompromised and in infants they can present serious problems78,79. Infection with persistent viruses have also been linked to autoimmune diseases, such as multiple sclerosis80,81, revealing their capacity to elicit aberrant immune programs. The specific mechanisms of the observed immune aberrancy, however, remains unclear.
Infection with persistent viruses has also been linked to broad phenotypic changes in T cells82,83. It is well established that CMV infection decreases the naïve T cell repertoire, as well as increases senescent memory cells, which has been reviewed extensively84. These senescent memory cells often have an impaired ability to divide upon stimulation. Interestingly, these broad phenotype changes have often been paralleled to changes that accumulate with age. In COVID-19, CMV infection has been highlighted as a potential risk factor for developing severe disease, specifically in non-geriatric patients (< 60 years)85. Such a phenomenon may be a consequence of CMV’s imprinting of an aged-like repertoire on younger immune systems. Interestingly, an independent study observed CD4+ and CD8+ T cells that could cross-react with both CMV and SARS-CoV-2 epitopes in SARS-CoV-2 unexposed individuals86. The authors also specifically delineated HLA-B*35:01-restricted CD8+ T cells binding to CMV pp65 and SARS-CoV-2 Spike epitopes in multiple individuals, rooted in a public TCR. Although the peptides share little homogeny (22%, Table 1), in silico structure analysis revealed a similar backbone conformation may be adopted in both pMHC structures. These cross-reactive T cells generally took either effector memory (EM) or effector memory RA (EMRA) phenotypes but did not activate during acute COVID-19. It is possible that these cross-reactive T cells bear a degree of immune senescence imparted by CMV infection that prevents them from participating in clearance of SARS-CoV-2. In support of this paradigm, another study observed a higher number of CD57+ (a marker indicative of cellular senescence in CD8+ T cells87) precursor SARS-CoV-2 specific CD8+ T cells in unexposed CMV seropositive versus seronegative individuals, mirroring that of the aged immune system88. A deeper understanding of how persistent viruses affect independent immune responses mechanistically could lead to better therapeutic interventions in cases of comorbidities.
T cell immunity to viral mutagenesis
Preservation of immunity to mutating viral strains is of great concern in vaccinology. Accumulation of mutations that abrogate binding of the repertoire can significantly cripple a vaccine’s effectiveness. A recent update in COVD-19 mRNA vaccines toward a bivalent formulation, containing both ancestral and omicron strains, is one such example that has been used in attempts to combat a drifting virus. Another prominent example is that of influenza vaccination, which requires annual updates to keep pace with a rapidly mutating virus. Due to the extensive length of production time in manufacturing seasonal influenza vaccines, variability in mutation predictions can lead to a wide range in efficacy. These inconsistencies highlight a critical need to deepen our knowledge of the immune response to mutagenesis. On the other end of the spectrum, HIV mutates at such a rapid rate that even seasonal vaccine updates would not be feasible. Such a phenomenon is one of the reasons that has made design of effective HIV vaccines an immense challenge. The following section will discuss mutation rates in SARS-CoV-2, influenza, and HIV and some of their immunological consequences.
Tracking propagation of mutations within the SARS-CoV-2 genome has thus been of great concern since its emergence in 2019. Due to proofreading machinery, SARS-CoV-2 accumulates mutations at a modest rate relative to other viruses – roughly 10−5 to 10−3 substitutions per base per infection cycle92. The sheer number of infections in the population (>760 million confirmed cases globally as of March 2023 according to the WHO health Emergency Dashboard); however, has led to many opportunities for the virus to mutate. As such, several variants of concern (VOCs) have emerged, often followed by cycles of increased infection rates. Several studies characterizing vaccine-induced antibody responses and therapeutic monoclonal antibodies have revealed a significant reduction in neutralization toward most VOCs, particularly toward the more recent Omicron lineages93,94. Newer bivalent formulations of the vaccine, which include Omicron spike glycoprotein-encoding mRNA, also do not appear to improve antibody neutralization compared to the old formulation95,96. In contrast, T cell immunity has been revealed to remain durable against Omicron lineages97–99, even contributing to preventing infection in some cases100.
A lack of mutations on immundominant epitopes may be a substantial reason that the T cell response to SARS-CoV-2 remains durable. To date, only five of the top twenty most cited immune epitope database (IEDB)101 SARS-CoV-2 spike-derived epitopes have been mutated in any dominant lineage – most of which are single amino acid point mutations. Building on this paradigm, a recent study revealed that a strong T cell response to these unmutated, immunodominant epitopes was associated with milder COVID-1938. One specific example of a mutated SARS-CoV-2 immunodominant epitope, however, is an HLA-A*24:02-restricted (A24/S448) epitope. In Delta and BA.4/5 lineages, an L452R substitution has been shown to increase infectivity89 (Table 1). Furthermore, CD8+ T cells from vaccinated donors respond poorly to the mutated epitope, suggesting lack of consistent cross-reactivity in the vaccine-induced repertoire102. It is thus possible that if SARS-CoV-2 continues to accumulate mutations, new variants will emerge that will not be durably cross-protected by previous T cell immunity.
Mutations in influenza A virus (IAV) are also a major concern. Although a CD8+ T cell response is not generally elicited by seasonal influenza vaccination, universal T cell epitopes have been characterized through immunopeptidomics59. Furthermore, as with SARS-CoV-2, T cell contribution during IAV infection is an important aspect of viral clearance103. T cells cross-reactive to several IAV strains have also been characterized structurally and via flow cytometry90 (Table 1), revealing a structural conservation of pMHC amongst the variants that likely underpins the high degree of observed cross-reactivity in the repertoire. Such a finding supports the argument for T cell consideration in seasonal influenza vaccination. In fact, significant effort has been directed toward the development of universal flu vaccines that could provide an immunological safeguard in the event of vaccine mismatches. Several clinical and pre-clinical studies involving T cell-based influenza vaccines have been reviewed recently104. Wide-spread implementation of safe and effective versions of these vaccines could lead to dramatic changes in the way we develop and think about vaccinology in general.
Even more so than SARS-CoV-2 and influenza, HIV provides an example that has been immensely challenging to design effective vaccines against. The high degree replicative errors within in the HIV genome makes it a particularly problematic example of mutation-based immune evasion. Unlike SARS-CoV-2, HIV contains quite promiscuous replication machinery. Being so, HIV-1 has been shown to accumulate on the order of 0.1 to 1 mutations per genome replication105–108. Such a high mutation burden is one of the major contributing factor of CD8+ T cell-mediated immune escape during chronic HIV infection109.
Although rare, there have been many cases of HIV-infected individuals, categorized as elite controllers and long-term non-progressors, that are capable of controlling virus below detection without therapeutic intervention for 10+ years110. An early connection between viral control and CD8+ T cells came from a study that linked long-term non-progressors to HLA-B*57:01111. The functional properties of CD8+ T cells have also been associated with viral control. Specifically, the ability to proliferate112 and kill113 upon stimulation with HIV antigens was consistently greater in controllers versus progressors. In contrast, cellular activation alone was not indicative of control as activated cells from progressors often exhibited exhausted phenotypes. Furthermore, HIV-specific CD8+ T cells remained exhausted after prolonged anti-retroviral treatment (ART)114. Although the imprinting of a defective CD8+ T cell phenotype is apparent, the mechanism of this imprinting remains unclear. Do these dysfunctional phenotypes in HIV mirror those observed in persistent viruses, other chronic infections, or uncontrolled cancer? Teasing apart the presence or lack of these inter-disease relationships could prove incredibly useful toward treatments of each of these contexts independently.
Comprehensive evaluation of the HIV specific repertoire remains a tremendous challenge due to the intra- and inter-infection diversity of the HIV genome. Typically, assays are limited to the response to a single reference strain, which may not be true to in vivo biology. However, strong and diverse responses to Gag have been affiliated with lower viral load, regardless of HLA background and mutation load115,116. Furthermore, modeling regions of Gag targeted by controllers versus progressors revealed structural biases indicative of mutational constraints117. Crystallographic analysis of the regions targeted by controllers also revealed a preference toward targets in interconnected regions of the protein118. Not only were mutations in these regions shown to impair viral fitness in general, but CD8+ T cells targeting these regions in elite controllers tended to target regions less mutated at TCR contact sites and MHC anchor residues. Cross-reactive, or promiscuous, public TCRs have also been associated with HIV control in an HLA-dependent manner119–122. Although cross-reactive TCRs alone do not discriminate progressors from non-progressors, these results further suggest certain repertoire characteristics may be favorable in some contexts. While it may not be a strict rule, certain HLA backgrounds may pre-dispose an individual to producing TCRs that target favorable regions of the HIV proteome and/or resist immune escape through TCR promiscuity. It is possible that TCR-intrinsic properties influence the functional outcomes shown to be indicative of viral control. For instance, persistent antigen exposure has been shown to lead to exhaustion in both chronic infection and cancer123. If HIV control leads to more “normal” antigen exposure in vivo, CD8+ T cells from non-progressors may be more inclined to take on functional memory, non-exhausted phenotypes that bear a greater potential to continue to control the virus. This phenomenon has been demonstrated to some degree in a small, high-risk cohort, the magnitude of the CD8+ T cell response was shown to correlate inversely with viremia in the early, hyperacute phase of HIV infection124. It is also possible that mutations on T cell targets may alter the quality of the TCR:pMHC contact, further leading to dysfunctional TCR signaling as has been seen in cancer models125.
Pathogenic-derived antigens in neurodegeneration
Recent studies have demonstrated that T cell infiltration and clonal expansion is enhanced in neurodegenerative disorders such as AD126–130, PD131–135, ALS136,137, and MS138–141. Although some of these disorders have T cell antigens that are more clearly defined such as in MS and PD, there is still a significant need for further investigating the antigenic source/breadth and the mechanisms in which they induce T cell activation.
While screening for T cell epitopes in pathogens is a well-defined task, screening for pathogen derived epitopes that mimic self-antigens is very challenging. Large scale epidemiological studies can often provide some clues. There is a growing body of evidence supporting the correlation between viral infections and cognitive disorders142,143. For example, AD pathogenesis has been associated with various viruses including herpes viruses, cytomegalovirus, HIV, Varicella zoster virus, EBV, and Hepatitis C143,144. Additionally, EBV infection is causally linked with MS as evidenced by an extensive longitudinal analysis of millions of US military personnel that were monitored for 20 years80. This study demonstrated that the risk of MS increased 32-fold after infection with EBV but was not increased after infection with other viruses such as CMV. Out of the 801 MS cases, only one individual was EBV-negative in the last collected sample. Another study analyzed 148 BCR sequences which were found in the CSF of MS patients and demonstrated molecular mimicry between the EBV transcription factor EBV nuclear antigen 1, EBNA1, and the CNS glial cell adhesion protein, GlialCAM81,145. Specifically, this group identified a monoclonal antibody clone that binds the MS-associated EBNA1 region, EBNA1AA386–405, which was then discovered to bind GlialCAM protein and phosphorylated peptide (pSer376 & pSer377) GlialCAMAA370–389 with high-affinity. The presence of EBV-infected memory B cells has various implications on the pathophysiology of MS and may impact how aberrantly activated disease-relevant or bystander T cells contribute the initiation or relapsing of disease146.
Using various T cell profiling strategies, Gate et al identified clonally expanded TEMRA (CD3+CD8+CD45RA+CD27−CD28−) cells enriched in the peripheral blood and CSF of patients with Alzheimer’s disease and mild cognitive impairment126. The group identified a TCRαβ clone within the CSF of a patient with MCI and AD with previously identified specificity to the Herpesviridae Epstein–Barr nuclear antigen 3 (EBNA3A, FLRGRAYGL). They further identified a TCR clone that had shared beta chain homology found between three AD patients (2 patients: CASSLAGGYNEQFF, 1 patient: CASSLGTGNNEQFF). They validated that the TCR found in the single patient (TCRα: CAASEGGFKTIF; TCRβ: CASSLGTGNNEQFF) was able to recognize the EBV trans-activator protein BZLF1 (RAKFKQLL) presented on HLA-B*08:01, while the specificity of the other TCR clone was undetermined due to the limited screen of 80 candidate peptides that was performed. As the authors note, this finding does not provide evidence of a causal link between EBV and AD but does suggest that T cells recognizing both self and non-self-antigens should be evaluated when investigating the contribution of T cells in neurodegeneration.
The Arlehamn and Sette groups evaluated T cell responses to common pathogenic antigens in both PD and AD compared to healthy controls but did not find significant differences using activation induced marker and peptide stimulation assays127,147. In their PD cohort, they evaluated T cells response to over 3000 viral and bacterial antigens across coronavirus, rhinovirus, respiratory syncytial virus, influenza, cytomegalovirus, pertussis, and tetanus across 19 patient and 20 healthy control PBMC samples.
Self-derived antigens in neurodegeneration
Some self-antigen screens have been performed on individuals who have MS, PD, AD, and other neurodegenerative disorders, but many of those screens have relied on functional stimulation assays which prevent detection of unmanipulated antigen-specific populations. For example, Arlehamn et al explored how tau pathology could influence the T cell landscape in PD, especially since aggregates of α-synuclein/tau oligomers are present in some patients148. They screened epitopes derived from tau and phosphorylated tau against T cells from control and PD patients using peptide stimulation of PBMC and measured cytokine production via ELISPOT. They identified that autoreactive T cell responses to tau were present at similar levels between PD patients and healthy controls. Interestingly, they also found that T cells responded more vigorously to tau than the PD-specific α-synuclein peptide. In a subsequent study, this group evaluated the frequency of T cells specific to amyloid precursor protein, amyloid beta, tau, α-synuclein, and TDP-43 in patients with AD and age-matched healthy controls using a similar peptide pool stimulation approach127. Similarly, no significant differences of cytokine response were found between AD and control.
Therefore, in evaluating T cell responses to self-antigens in neurodegeneration, one cannot assume that central tolerance is complete. Likewise, another study by Sabatino Jr et al demonstrated that there are similar frequencies of myelin-specific T cells in MS and healthy control, but the myelin-specific T cells in MS exhibit a non-naïve, antigen-experienced phenotype139. The group surveyed the landscape of myelin-reactive T cells in the PBMC of MS patients across five validated epitopes and found similar frequencies of myelin-specific T cells in MS and healthy control. However, the antigen-specific T cells found in MS were non-naïve, expressed CD20, and were significantly reduced in patients treated with anti-CD20 mAb therapy, suggesting prior antigen experience. These studies suggest that it is not always sufficient to only identify the presence or frequency of autoreactive T cells, but to fully characterize their specificity, phenotype, and functional capacity thoroughly to be able to understand which features are most implicated in disease.
In PD, the presence of self-reactive T cells toward epitopes derived from the pathological protein α-synuclein has a much stronger implication. A series of studies conducted by Arlehamn and Sette identified that more pronounced cytokine production was evident in PD compared to healthy controls in response to stimulation with several antigenic regions of α-synuclein (Y39: α-syn31–45 GKTKEGVLYVGSKTK, α-syn32–46 KTKEGVLYVGSKTKE and phosphorylated S129: α-syn116–130 MPVDPDNEAYEMPSE, α-syn121–135 DNEAYEMPSEEGYQD, α-syn126–140 EMPSEEGYQDYEPEA)131. A longitudinal case study identified α-synuclein specific (α-syn61–75 EQVTNVGGAVVTGVT) T cells in the peripheral blood of a motor PD patient years prior to their diagnosis, with the magnitude of the response waning after diagnosis132. Recently, these groups evaluated the TCR repertoire of α-synuclein-specific T cells in six PD patients and found that the repertoire was as diverse as the repertoire to antigens derived from Pertussis149. No public α-synuclein TCR was identified across the patients, though the patients were not HLA matched.
Campisi et al discovered highly expanded and activated CD8+ TEMRA cells in the PBMC of patients with ALS-4 and in the spinal cord, brain, and PBMC of mice with the ALS4-causative Senataxin L389S mutation136. Interestingly, these mice were able to control an induced high-grade glioma with high levels of activated CD8+ T cell infiltration, but not melanoma, suggesting an immune response directed against self-antigens of CNS origin. The clonal T cell sequences were also cross-referenced to known pathogenic antigens, and no overlapping sequences were identified. Additionally, control and ALS-4 PBMCs were stimulated with pools of self-peptide (TDP-43, Senataxin) or pathogen-derived peptides and IFNγ concentration was measured, but no difference between control and ALS-4 was identified. This result highlights the challenge of identifying and screening T cell antigens, even those which are strongly implicated in disease.
TCR cross-reactivities in neurodegeneration
Krishnamoorthy et al observed a paradoxical result in which spontaneous optic neuritis developed in TCR 2D2 MOG-deficient (MOG−/−) transgenic mice which harbored TCRs specific to MOG35–55 while IgHMOG × MOG−/− mice with B cells specific for MOG remained healthy91. They identified that the 2D2 transgenic T cells were cross-reactive to a neuronal cytoskeletal self-antigen, NF-M18–30 which contained homology to the core residues of the MOG35–55, but not to NF-M225–237 (Table 1). Additionally, they determined that NF-M18–30 can be targeted by polyclonal T cells isolated from MOG35–55-specific T cells from C57BL/6 mice, indicating that this cross-reactive pair is not limited to a response by a single T cell clone.
In 1991, Brian Evavold and Paul Allen introduced altered peptide ligands (APLs) by demonstrating that the TCR can have differential signaling if its cognate epitope is conservatively altered with a single amino acid mutation150. A subsequent study by Dresseln et al aimed to evaluate the functional role of APLs in the context of peptides relevant in multiple sclerosis151. They screened ten variations of the peptide PLP80–88 (FLYGALLLA), derived from myelin proteolipid protein for HLA-A2 binding along with cytotoxicity, cytokine secretion, and proliferative capacity against several T cell clones. Most of the ALPs had an HLA-A2 binding capacity (IC50) close to the wildtype (2.6 nM) with the exception of FLAGALLLA (599 nM) and FLYAALLLA (17,177 nM). All altered peptides except FLAGALLLA were able to induce cytotoxicity by at least one of the PLP80–88-specific T cell clones that were evaluated. Additionally, the L ➔ A mutation in position 87, FLYGALLAA, was able to act as a superagonist capable of inducing half-maximal cytotoxic, cytokine, and proliferative response with 100-fold less concentration compared to the wildtype.
APLs which compete for TCR binding but do not lead to full cellular activation have demonstrated efficacy in treating and reversing EAE through their capacity to induce a TH1 to TH2 phenotype switch in myelin-specific T cells152. However, in clinical trials, this approach has faced significant challenges and has highlighted the complexity of the T cell response. A large double-blind phase II trial led by Neurocrine Biosciences evaluated an APL designed from the immunodominant myelin basic protein epitope, MBP83−99 (NBI-5788), consisting of the following substitutions: D-Ala83-Lys84-Leu89-Ala91 (ENPVVHFFKNIVTPRTP ➔ AKPVVHLFANIVTPRTP). The APL was administered at various doses, and although there was no difference of the relapse rate between the APL and placebo group, there were smaller and fewer lesions in patients which received the lowest dose. However, the trial was suspended due to 13/142 patients developing immediate-type hypersensitivity and generating anti-NBI-5788 antibodies which could cross-react with the native peptide152–154. A smaller trial which included only eight patients evaluated the same APL at the higher dose level reported a strongly immunogenic response that led to the expansion of cross-reactive T cells for the APL and native protein. These trials suggest that although an APL may be capable of inducing anergy or an anti-inflammatory phenotype switch in some T cell clones, it could lead to activation of other clonotypes152,153,155.
Cancer antigens
Leveraging the T cell ability to specifically recognize and destroy cancer is fundamental to the study of cancer immunology and the future progress of cancer immunotherapy. Cancer antigens can be categorized into two groups, tumor associated antigens (TAAs) and tumor specific antigens. Tumor associated antigens are not specific to tumors. They are expressed in healthy tissue but with an elevated expression in the tumor. For example, the first human tumor antigen identified to be recognized by T cells, mucin156, is widely expressed on many types of cancers. Due to a lack of expression in healthy cells, tumor specific antigens provide great on-target specificity with minimum side effects. Tumor specific antigens can be categorized into four subclasses. Cancer testis antigens (CTA), also known as cancer germline antigens, are expressed in the embryonic stage and testis tissue but are epigenetically silenced in adult peripheral tissues. Because their expression at both RNA and protein levels have been detected in many types of cancers157,158, these antigens are valuable candidate for cancer immunotherapy. However, not all CTAs are the same. In a recent clinical trial, autologous TCRs recognizing a shared epitope by several CTAs, MAGE-A3/A9/A12, caused severe neurological toxicity because unexpected expression of MAGE-A12 in human brains159, highlighting the importance of selecting CTAs and understanding their individual tissue expression level for immunotherapy. The second class of tumor specific antigens is neo-antigens. Neo-antigens are mutated variants of self-proteins that are exclusively expressed by tumor cells (extensively reviewed in refs160–164). Depending on the self-proteins, neo-antigens can also be categorized as driver mutation-derived neo-antigens that are shared among different types of cancers or patient private mutations derived neo-antigens that are personalized to each patient. The third class of tumor specific antigens are antigens generated from non-protein-coding regions of DNA that are often called cryptic antigens. One example are peptides derived from alternative splicing or intronic retention or other post-transcriptional events165. The fourth class of tumor specific antigens are viral antigens expressed by cancers resulted from viral infections, such as hepatitis B and C viral antigens in hepatitis B- and C-related hepatocellular carcinomas and human papillomavirus in cervical cancer. In addition to their unique tumor expression profile, neo-antigens and viral antigens are truly foreign that should retain high affinity TCRs from the thymic selection process166.
Cancer antigen specific T cells
Initial cancer antigen specific T cell identification efforts primarily used limiting dilution analysis167. Using in vitro culture and cytotoxicity assays on autologous tumor cells, it has been shown that tumor reactive T cells can be identified168. Using TCR V region antibodies, it was possible to track antigen specific T cell repertoire169 and measure their clonal size dynamics170 and phenotypes171 through vaccination. By combining flow cytometry sorting and PCR, analyzing TCR gene usage172 was made possible for a polyclonal population of T cells responding to mouse syngeneic tumor cells transfected with a model peptide. A breakthrough came when John Alterman, Mark Davis, and colleagues developed pMHC tetramers173 which can overcome the fast off-rate of low-affinity TCR-pMHC interactions. Using pMHC tetramers made with the TAAs MART-1, gp100, and tyrosinase peptides for melanoma174 and PR1 for myeloid leukemia175, they showed that functional tumor specific T cells can be isolated from CD8 T cell lines generated from healthy donors’ PBMCs which could be exempt from the exhaustion program in the tumor microenvironment. Around the same time, using TAAs in melanoma, Labarriere et al176, and Romero et al177, independently identified tumor antigen specific T cells in metastatic lymph nodes of melanoma patients. Lee et al even analyzed the cytotoxicity of the T cells isolated from melanoma patients’ PBMCs178 and showed that they were functionally unresponsive compared with EBV or CMV specific T cells analyzed ex vivo. This is the first time that tumor antigen-specific T cell phenotype and function could be analyzed in their “native” state without prior stimulation.
Many studies have focused on neo-antigens because they are uniquely expressed in cancer tissue. This property enhances the on-target specificity of neo-antigen specific T cells while reducing the off-target toxicity. Since the discovery of the first T cell cognate neo-antigens in mice179 and humans180, large scale neo-antigen discovery have been aided by the development of next-generation sequencing (NGS) technologies and their applications in analyzing cancer genomes181–183. However, the discovery of neo-antigen specific T cells in those early days was only attainable for a few patients and was largely dependent on the ability to successfully generate in vitro T cell clones161. Robbins et al demonstrated for the first time that a patient autologous tumor infiltrating lymphocyte (TIL) derived T cell clone was able to recognize a neo-antigen184. The first attempt to exhaustively look for neo-antigens and autologous cognate T cells was done by Lennerz et al185 prior to the development of NGS. Using a cDNA expression screen method, they discovered five neo-antigens and cognate T cell clones from the patient’s peripheral blood in addition to three cancer associated antigens. This study demonstrated that the immune system has the capacity to target multiple cancer antigens, including multiple neo-antigens.
Aided by a peptide exchange method to quickly generate new pMHC species186,187, Newell et al188 and Hadrup et al189 independently developed a combinatorial method to increase the number of pMHCs that can be paralleled in the analysis. Using this method, Hadrup et al performed a screen of 22 pMHCs, derived from four melanoma associated antigens, in PMBCs from 28 HLA matched melanoma patients. A total of 24 T cell responses targeting eight epitopes were detected, five of which are previously unknown T cell epitopes. Further increases of the number of pMHC tetramer species that can be multiplexed arrived when the mass cytometry technology190 became available. Subsequently, Newell et al combined mass cytometry with combinatorial tetramer stain and significantly expanded the number of multiplexed pMHC tetramers to over a hundred191. This technology opened the possibility of predicting and screening a large number of new T cell epitopes. Using mass cytometry, Simoni et al192 screened 1091 putative neoantigens, 123 TAAs, and 46 cancer-unrelated epitopes in TILs derived from 24 patients with various types of cancers. Cognate TILs were detected for neoantigen epitopes but not any of the TAAs screened. Unexpectedly, they discovered a large population of common viral epitope specific TILs. These bystander CD8+ TILs lack CD39 expression compared to tumor antigen specific TILs, suggesting that tumor antigen specific TILs may have a distinct phenotype that could be further leveraged for their identification. Independently, Duhen et al193 showed that co-expression of CD39+ and CD103+ identifies tumor-reactive CD8+ TILs in six different types of cancers, suggesting the value of focusing on a subset of TILs for prognosis during immunotherapy and identifying tumor antigen-specific T cells. Similar to Simoni et al192, Scheper et al194 also found that only about 10% of CD8+ TILs recognize autologous tumor. Therefore, the antigen landscape of TILs must be much more complex. Understanding the complexity of antigen specific T cells in tumor microenvironment could motivate new therapeutic development.
In addition to identifying bystander viral specific T cells in the TILs, Rosato et al195 showed that injecting viral peptides into mouse tumors triggered antigen presentation and cytotoxicity in the tumor and, most strikingly, rendered PD-L1 blockade resistant mouse tumors susceptible to the treatment. Viral peptide-treated ex vivo human tumors recapitulated immune activation gene profiles observed in mice. Similarly, virus specific CD8+ T cells also populate mouse and human glioblastomas, which are one of the most aggressive and treatment-resistant cancers196. Thus, these studies suggest that intratumoral delivery of viral peptide triggers local immune activation and activating viral bystander cells in TILs represents an alternative or complementary approach to tumor antigen specific TIL activation in cancer immunotherapy.
With single cell RNA sequencing (scRNA-seq) becoming more accessible, it is possible to use TCR sequences as T cell IDs to link transcriptome data acquired directly from TILs to antigen specificity acquired from ex vivo expanded TILs. Oliveira et al197 applied this strategy on a cohort of melanoma patients that received either neo-antigen vaccine or immune checkpoint blockade therapies. In addition to neo-antigen and common viral antigen specific TILs, they discovered TAA specific TILs. Non-tumor-reactive T cells exhibited a non-exhausted memory phenotype, whereas both TAA and neo-antigen reactive TILs displayed an exhausted state. Using a similar approach, Caushi et al198 examined the clonality and activation status of neo-antigen and viral-antigen specific TILs and in a cohort of non-small cell lung cancer patients that received anti-PD-1 treatment before tumor resection surgery. Similar to the other two studies192,197, neo-antigen specific TILs exhibited an incompletely activated cytolytic program. Although neo-antigen specific T cells were found in both anti-PD-1 responders and non-responders, the ones found in non-responders showed lower ligand-dependent signaling, coordinately upregulated checkpoints, and other features that inhibit T cell activation. In a separate study, Lowery et al199 used 55 TCR sequences from previously generated neo-antigen reactive T cell clones to track T cells in scRNA-seq data generated using archives of metastatic tumor samples. Using these signature TCRs, they were able to identify a subset of T cells that enriched with neo-antigen specific TCR. This allowed them to focus on a much smaller set of candidate TCRs and validate the neo-antigen targets in half of the predicted TCRs. These two studies opened the possibility of using neo-antigen specific T cell associated transcriptional program to track responses and predict new neo-antigen targeting TCRs. While this approach has been pursued mainly in cancer, possibly because of the unique transcriptional program of the tumor infiltrating T cells, it may be valuable to apply it to infection and neurodegenerative diseases. Recently, Puig-Saus et al200 performed the largest neo-antigen specific T cell screen ever conducted for seven anti-PD-1 responders and four non-responders using DNA-barcoded pMHC multimers201–203 on PBMCs and TIL cultures. Although neo-antigen cognate T cells were detected from both responders and non-responders, the ones from responders have more clonal expansion and showed up in multiple samples collected at different timepoints compared to the ones from non-responders.
Most of these attempts to identify T cell cognate neo-antigens focused on tumor mutational burden high (TMB-H) cancers204,205 with a hypothesis that TMB-H cancers will induce more neo-antigen recognition by T cells163. However, various attempts in different types of cancers, including the ones discussed above, showed that the T cell cognate neo-antigen discover rate remains low, about 0–4%. The healthy status of the TILs, bias introduced by in vitro expansion, and various cancer immune evasion mechanisms, including HLA loss-of-heterozygosity, disruption to antigen presentation, and repression of neoantigen expression206, all contribute to this low discovery rate. Zamora et al207 took a different approach and found that 86% of the neo-antigens and 68% of the neo-peptides (multiple peptides containing the same mutation) are recognized by patients’ autologous CD8+ T cells in pediatric acute lymphoblastic leukemia (ALL) that is on the extremely low end of the TMB spectrum163,205. This study suggested that having a few mutations may allow the immune system to focus on its repertoire and other recourse to mount a better immune response. Different tumor types could give rise to this difference. In addition, this study suggested that there might be fundamental differences between how pediatric immune systems recognize antigens compared to that of adults, which should be interesting to test in adult ALL patients.
McGranahan et al208 showed that in both non–small cell lung cancer and melanoma, sensitivity to PD-1 and CTLA-4 blockade treatment was enhanced in patients who had more neo-antigens present in all tumor cells compared to patients who had neo-antigens in only a fraction of tumor cells. Steven Rosenberg’s group showed in a series of papers that hotspot driver mutation can be targeted in adoptive cell transfer (ACT) therapy209. However, targeting a single neo-antigen only induced short-term clinical benefits and the cancer eventually evaded the immune system by downregulating the neo-antigen presenting HLA allele210. In a later study, they showed that targeting multiple neo-antigens could result in complete durable regression in metastatic breast cancer. These studies highlight the importance of understanding the neo-antigen expression heterogeneity in selecting treatment targets to overcome immune evasion.
In addition to their abundance in spontaneous cancers, viral peptide specific T cells are a major therapeutic target in virus-induced cancers. Combining mass scRNA-seq with the use of a large panel of viral antigens, TAAs, bystander epitopes, and neo-antigens, Cheng et al211 examined the antigen specificities of hepatocellular carcinoma (HCC) TILs, revealing that hepatitis B virus specific T cells exhibited a resident memory phenotype and transcriptional program. These cells were clonally expanded but were PD-1loTOXlo and not terminally exhausted. Patients with these cells infiltrating to tumor had a longer-term relapse-free survival. Similarly, Eberhardt et al212 screened and tracked human papillomavirus specific T cells in head and neck squamous cell carcinoma (HNSCC). TCR tracking and scRNA-seq analyses suggested hypothetical differentiation trajectory from TCF-1+PD-1+ stem-like subset to transitory to terminally differentiated cells. In vitro peptide stimulation also confirmed their proliferation and differentiation capacity. Although both studies focused on virus-induced cancers, different viral infections, tumor microenvironments, or both could give rise to different TIL subsets that inform different treatment options. While immune checkpoint blockade therapies had poor responses in HCC213, they have gained FDA approval for recurrent/metastatic HNSCC214,215. Both studies identified additional viral peptides that could be leveraged in the design of preventive and therapeutic vaccines that could be used in conjunction with other therapies.
TCR cross-reactivities in cancer
Although TCRs can specifically recognize tumor antigens, their cross-reactivity to other antigens have caused severe problems. One such example is an affinity enhanced TCR to its original antigen, CTA MAGE-A3, that caused death of two patients in a clinical trial. The fatalities were later linked to MAGE-A3’s cross-reactivity to a peptide derived from the muscle protein Titin216,217. This has motivated alternative ways to fine tune TCR affinity and validate them in pre-clinical models218. Recent biophysical studies showed that “catch bonds”, initially discovered in selectins50 and integrins51 and thought unique to adhesion molecules219, existed in TCRs52. Chao et al. showed that high-affinity TCRs, measured by the traditional surface plasmon resonance, are often cross-reactive to self-antigens, however, low-affinity TCRs often lack functional efficacy. They demonstrated that it is possible to engineer TCRs with a low affinity to cancer antigen but have a strong cytotoxicity if the TCRs have a catch bond property. These TCRs are enhanced with on-target cancer antigen recognition but without cross-reactivity to off-target self-antigens53. This study provides a new avenue for engineering antigen specific TCRs for therapeutic development that can be applied to many other fields, such as infections and neurodegenerative diseases.
Other studies have explored possibilities of neo-antigen cognate T cells cross-reacting with wildtype proteins. Zhang et al showed that this type of T cell is readily detected in healthy individuals by using a large number of DNA-barcoded pMHC tetramer202. They also showed that these cells can be functionally validated through in vitro stimulation. By combining comprehensive peptide single position permutation experiments with informatics approach on a NY-ESO-1 peptide, Karapetyan et al220 validated 7 out of 12 highly scored cross-reactive peptide predicted from human proteome, including one that was 7 amino acid changes from the original nonomer peptide. These studies highlighted the complexity that TCR-antigen interaction spaces are and the importance of understanding the fundamental rule about TCR-antigen recognition has on cancer immunotherapy.
Informatic T cell epitope prediction
Computational prediction of TCR specificity can have immense utility in furthering our understanding of systems immunology and would lead to breakthroughs in translational immunotherapies. Recent reviews thoroughly highlight the various in silico modeling approaches aimed at predicting peptide-MHC binding, cross-reactivity, immunogenicity, and TCR-pMHC interaction221–226. Despite recent advances in both high-throughput TCR-antigen discovery and machine learning approaches, there exist significant challenges that need to be addressed. The fundamental limitation in most of these computational approaches can be attributed to the lack of experimentally validated, publicly available TCR-antigen pairs221,224. In fact, 97% of TCR-antigen pairs are those which are of viral origin and TCRs specific to a set of 100 antigens make up about 70% of the currently existing data221. Additional limitations include the lack of paired alpha-beta chains, HLA bias, lack of true negative datasets, and binary representation of the data (binder vs. non-binder)221,223,224.
Many groups are actively developing novel strategies to overcome these limitations. Gao et al. developed Pan-Peptide Meta Learning (PanPep)227,228, a framework which combines concepts of meta-learning and the neural Turing machine to address the challenge of the long-tail distribution that is characteristic in TCR-epitope data. Their model is constructed for three levels of predictions based on the amount of known TCRs for a given peptide: none (zero-shot), few (few-shot), and majority. PanPep significantly outperformed existing tools in the zero-shot and few-shot predictions. However, PanPep only considers the CDR3b chain and does not consider other relevant information such as alpha chain, HLA type, or 3D structure. Bradley229 aimed to predict TCR-epitope interaction through structure-based analysis of the TCR:peptide-MHC complex by utilizing a custom version of the neural network predictor AlphaFold. Their pipeline, AlphaFold TCR, aims to select the correct target peptide from a small set of candidate peptides. This work provides a good proof-of-concept and highlights the utility in incorporating structure-based information in TCR-epitope predictions. Other groups are evaluating novel ways of the initial embedding of the TCR sequence to numeric representations. Zhang et al. propose catELMo230, a bi-directional context-aware amino acid embedding model that treats amino acids as words and sequences as sentences. They demonstrate that their embedding approach can outperform other widely used approaches such as BLOSUM62 and BERT-based embedding models. Other groups are focused on gathering large datasets or generating databases to have more robust and easy-to-access training sets. Zhou et al. constructed NeoTCR231, an immunoinformatic database consisting of publicly available neoantigen-specific TCR sequences across 18 cancer subtypes. This further highlights the need for a unified, well-annotated, and reliable TCR-epitope database.
Concluding remarks
The complex landscape of T cell and antigen interactions are critical to disease initiation and pathogenesis as well as diagnosis and therapeutic development. Because of the enormous impact both scientifically and therapeutically, waves of technological developments have advanced the field significantly in the last 30 years. Recent innovations make the exploration of the intricate interactions in the mesh network of TCR and antigen repertoires possible. Accompanying this is the exponential increase of high-dimensional data that link TCR-antigen interactions with T cell states. With groundbreaking computational tools that could enable the prediction of TCR antigen specificity and novel approaches for TCR engineering, the future of antigen-specific TCR-based disease diagnosis and therapeutics is incredibly bright. A deep understanding of the antigen specific immune response could unlock paradigm-shifting therapeutic potential, allowing us to harness its vast power for both protection from, and treatments against an immense array of diseases, from infections to neurodegenerative diseases to cancer.
ACKNOWLEDGMENTS
This work is in part supported by the US National Institutes of Health grants R33CA225539 and R33CA256086, Cancer Research Institute grant 10081684, and Chan Zuckerberg Initiative grant 2021–005904 to N.J.
Footnotes
All authors declare that they have no conflicts of financial interest
References:
- 1.Hedrick SM, Cohen DI, Nielsen EA, Davis MM. Isolation of cDNA clones encoding T cell-specific membrane-associated proteins. Nature 1984;308(5955):149–153. [DOI] [PubMed] [Google Scholar]
- 2.Yanagi Y, Yoshikai Y, Leggett K, Clark SP, Aleksander I, Mak TW. A human T cell-specific cDNA clone encodes a protein having extensive homology to immunoglobulin chains. Nature 1984;308(5955):145–149. [DOI] [PubMed] [Google Scholar]
- 3.Bradley P, Thomas PG. Using T Cell Receptor Repertoires to Understand the Principles of Adaptive Immune Recognition. Annu Rev Immunol 2019;37:547–570. [DOI] [PubMed] [Google Scholar]
- 4.Dupic T, Marcou Q, Walczak AM, Mora T. Genesis of the alphabeta T-cell receptor. PLoS Comput Biol 2019;15(3):e1006874. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 5.Jenkins MK, Chu HH, McLachlan JB, Moon JJ. On the composition of the preimmune repertoire of T cells specific for Peptide-major histocompatibility complex ligands. Annu Rev Immunol 2010;28:275–294. [DOI] [PubMed] [Google Scholar]
- 6.Masopust D, Vezys V, Marzo AL, Lefrancois L. Preferential localization of effector memory cells in nonlymphoid tissue. Science 2001;291(5512):2413–2417. [DOI] [PubMed] [Google Scholar]
- 7.Reinhardt RL, Khoruts A, Merica R, Zell T, Jenkins MK. Visualizing the generation of memory CD4 T cells in the whole body. Nature 2001;410(6824):101–105. [DOI] [PubMed] [Google Scholar]
- 8.Steinert EM, Schenkel JM, Fraser KA, et al. Quantifying Memory CD8 T Cells Reveals Regionalization of Immunosurveillance. Cell 2015;161(4):737–749. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 9.Sender R, Fuchs S, Milo R. Revised Estimates for the Number of Human and Bacteria Cells in the Body. PLoS Biol 2016;14(8):e1002533. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 10.Mu L, Kang JH, Olcum S, et al. Mass measurements during lymphocytic leukemia cell polyploidization decouple cell cycle- and cell size-dependent growth. Proc Natl Acad Sci U S A 2020;117(27):15659–15665. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 11.Mason D A very high level of crossreactivity is an essential feature of the T-cell receptor. Immunol Today 1998;19(9):395–404. [DOI] [PubMed] [Google Scholar]
- 12.Moon JJ, Chu HH, Pepper M, et al. Naive CD4(+) T cell frequency varies for different epitopes and predicts repertoire diversity and response magnitude. Immunity 2007;27(2):203–213. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 13.Chu HH, Moon JJ, Takada K, et al. Positive selection optimizes the number and function of MHCII-restricted CD4+ T cell clones in the naive polyclonal repertoire. Proc Natl Acad Sci U S A 2009;106(27):11241–11245. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 14.Obar JJ, Khanna KM, Lefrancois L. Endogenous naive CD8+ T cell precursor frequency regulates primary and memory responses to infection. Immunity 2008;28(6):859–869. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 15.Kotturi MF, Scott I, Wolfe T, et al. Naive precursor frequencies and MHC binding rather than the degree of epitope diversity shape CD8+ T cell immunodominance. J Immunol 2008;181(3):2124–2133. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 16.Haluszczak C, Akue AD, Hamilton SE, et al. The antigen-specific CD8+ T cell repertoire in unimmunized mice includes memory phenotype cells bearing markers of homeostatic expansion. J Exp Med 2009;206(2):435–448. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 17.Su LF, Kidd BA, Han A, Kotzin JJ, Davis MM. Virus-specific CD4(+) memory-phenotype T cells are abundant in unexposed adults. Immunity 2013;38(2):373–383. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 18.Yu W, Jiang N, Ebert PJ, et al. Clonal Deletion Prunes but Does Not Eliminate Self-Specific alphabeta CD8(+) T Lymphocytes. Immunity 2015;42(5):929–941. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 19.Shimonkevitz R, Colon S, Kappler JW, Marrack P, Grey HM. Antigen recognition by H-2-restricted T cells. II. A tryptic ovalbumin peptide that substitutes for processed antigen. J Immunol 1984;133(4):2067–2074. [PubMed] [Google Scholar]
- 20.Townsend AR, Bastin J, Gould K, Brownlee GG. Cytotoxic T lymphocytes recognize influenza haemagglutinin that lacks a signal sequence. Nature 1986;324(6097):575–577. [DOI] [PubMed] [Google Scholar]
- 21.Buus S, Sette A, Colon SM, Jenis DM, Grey HM. Isolation and characterization of antigen-Ia complexes involved in T cell recognition. Cell 1986;47(6):1071–1077. [DOI] [PubMed] [Google Scholar]
- 22.Buus S, Colon S, Smith C, Freed JH, Miles C, Grey HM. Interaction between a “processed” ovalbumin peptide and Ia molecules. Proc Natl Acad Sci U S A 1986;83(11):3968–3971. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 23.Babbitt BP, Matsueda G, Haber E, Unanue ER, Allen PM. Antigenic competition at the level of peptide-Ia binding. Proc Natl Acad Sci U S A 1986;83(12):4509–4513. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 24.Bjorkman PJ, Saper MA, Samraoui B, Bennett WS, Strominger JL, Wiley DC. The foreign antigen binding site and T cell recognition regions of class I histocompatibility antigens. Nature 1987;329(6139):512–518. [DOI] [PubMed] [Google Scholar]
- 25.Carbone FR, Moore MW, Sheil JM, Bevan MJ. Induction of cytotoxic T lymphocytes by primary in vitro stimulation with peptides. J Exp Med 1988;167(6):1767–1779. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 26.Rudensky Y, Preston-Hurlburt P, Hong SC, Barlow A, Janeway CA Jr., Sequence analysis of peptides bound to MHC class II molecules. Nature 1991;353(6345):622–627. [DOI] [PubMed] [Google Scholar]
- 27.Makarov A Electrostatic axially harmonic orbital trapping: a high-performance technique of mass analysis. Analytical chemistry 2000;72(6):1156–1162. [DOI] [PubMed] [Google Scholar]
- 28.Laumont CM, Daouda T, Laverdure JP, et al. Global proteogenomic analysis of human MHC class I-associated peptides derived from non-canonical reading frames. Nat Commun 2016;7:10238. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 29.Delong T, Wiles TA, Baker RL, et al. Pathogenic CD4 T cells in type 1 diabetes recognize epitopes formed by peptide fusion. Science 2016;351(6274):711–714. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 30.Slavov N Single-cell protein analysis by mass spectrometry. Curr Opin Chem Biol 2021;60:1–9. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 31.Alfaro JA, Bohlander P, Dai M, et al. The emerging landscape of single-molecule protein sequencing technologies. Nat Methods 2021;18(6):604–617. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 32.Rodenko B, Toebes M, Celie PH, Perrakis A, Schumacher TN, Ovaa H. Class I major histocompatibility complexes loaded by a periodate trigger. J Am Chem Soc 2009;131(34):12305–12313. [DOI] [PubMed] [Google Scholar]
- 33.Saini SK, Schuster H, Ramnarayan VR, Rammensee HG, Stevanovic S, Springer S. Dipeptides catalyze rapid peptide exchange on MHC class I molecules. Proc Natl Acad Sci U S A 2015;112(1):202–207. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 34.Luimstra JJ, Garstka MA, Roex MCJ, et al. A flexible MHC class I multimer loading system for large-scale detection of antigen-specific T cells. J Exp Med 2018;215(5):1493–1504. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 35.Overall SA, Toor JS, Hao S, et al. High throughput pMHC-I tetramer library production using chaperone-mediated peptide exchange. Nat Commun 2020;11(1):1909. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 36.Sun Y, Young MC, Woodward CH, et al. Universal open MHC-I molecules for rapid peptide loading and enhanced complex stability across HLA allotypes. bioRxiv 2023. [DOI] [PMC free article] [PubMed]
- 37.Huang J, Zeng X, Sigal N, et al. Detection, phenotyping, and quantification of antigen-specific T cells using a peptide-MHC dodecamer. Proc Natl Acad Sci U S A 2016;113(13):E1890–1897. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 38.Mallajosyula V, Ganjavi C, Chakraborty S, et al. CD8(+) T cells specific for conserved coronavirus epitopes correlate with milder disease in COVID-19 patients. Sci Immunol 2021;6(61). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 39.Sugata K, Matsunaga Y, Yamashita Y, et al. Affinity-matured HLA class II dimers for robust staining of antigen-specific CD4(+) T cells. Nat Biotechnol 2021;39(8):958–967. [DOI] [PubMed] [Google Scholar]
- 40.Dileepan T, Malhotra D, Kotov DI, et al. MHC class II tetramers engineered for enhanced binding to CD4 improve detection of antigen-specific T cells. Nat Biotechnol 2021;39(8):943–948. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 41.Joglekar AV, Li G. T cell antigen discovery. Nat Methods 2021;18(8):873–880. [DOI] [PubMed] [Google Scholar]
- 42.Dahotre SN, Chang YM, Romanov AM, Kwong GA. DNA-Barcoded pMHC Tetramers for Detection of Single Antigen-Specific T Cells by Digital PCR. Analytical chemistry 2019;91(4):2695–2700. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 43.Ma KY, Schonnesen AA, He C, et al. High-throughput and high-dimensional single-cell analysis of antigen-specific CD8(+) T cells. Nat Immunol 2021;22(12):1590–1598. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 44.Liu Z, Li JP, Chen M, et al. Detecting Tumor Antigen-Specific T Cells via Interaction-Dependent Fucosyl-Biotinylation. Cell 2020;183(4):1117–1133 e1119. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 45.Arnaud M, Chiffelle J, Genolet R, et al. Sensitive identification of neoantigens and cognate TCRs in human solid tumors. Nat Biotechnol 2022;40(5):656–660. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 46.Guo XJ, Elledge SJ. V-CARMA: A tool for the detection and modification of antigen-specific T cells. Proc Natl Acad Sci U S A 2022;119(4). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 47.Yu B, Shi Q, Belk JA, et al. Engineered cell entry links receptor biology with single-cell genomics. Cell 2022;185(26):4904–4920 e4922. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 48.Bell GI. Models for the specific adhesion of cells to cells. Science 1978;200(4342):618–627. [DOI] [PubMed] [Google Scholar]
- 49.Dembo M, Torney DC, Saxman K, Hammer D. The reaction-limited kinetics of membrane-to-surface adhesion and detachment. Proc R Soc Lond B Biol Sci 1988;234(1274):55–83. [DOI] [PubMed] [Google Scholar]
- 50.Marshall BT, Long M, Piper JW, Yago T, McEver RP, Zhu C. Direct observation of catch bonds involving cell-adhesion molecules. Nature 2003;423(6936):190–193. [DOI] [PubMed] [Google Scholar]
- 51.Kong F, Garcia AJ, Mould AP, Humphries MJ, Zhu C. Demonstration of catch bonds between an integrin and its ligand. J Cell Biol 2009;185(7):1275–1284. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 52.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]
- 53.Zhao X, Kolawole EM, Chan W, et al. Tuning T cell receptor sensitivity through catch bond engineering. Science 2022;376(6589):eabl5282. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 54.Klinger M, Pepin F, Wilkins J, et al. Multiplex Identification of Antigen-Specific T Cell Receptors Using a Combination of Immune Assays and Immune Receptor Sequencing. PloS one 2015;10(10):e0141561. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 55.Chng MHY, Lim MQ, Rouers A, et al. Large-Scale HLA Tetramer Tracking of T Cells during Dengue Infection Reveals Broad Acute Activation and Differentiation into Two Memory Cell Fates. Immunity 2019;51(6):1119–1135 e1115. [DOI] [PubMed] [Google Scholar]
- 56.Cheng Y, Zhu YO, Becht E, et al. Multifactorial heterogeneity of virus-specific T cells and association with the progression of human chronic hepatitis B infection. Sci Immunol 2019;4(32). [DOI] [PubMed] [Google Scholar]
- 57.Saini SK, Hersby DS, Tamhane T, et al. SARS-CoV-2 genome-wide T cell epitope mapping reveals immunodominance and substantial CD8(+) T cell activation in COVID-19 patients. Sci Immunol 2021;6(58). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 58.Francis JM, Leistritz-Edwards D, Dunn A, et al. Allelic variation in class I HLA determines CD8(+) T cell repertoire shape and cross-reactive memory responses to SARS-CoV-2. Sci Immunol 2022;7(67):eabk3070. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 59.Koutsakos M, Illing PT, Nguyen THO, et al. Human CD8(+) T cell cross-reactivity across influenza A, B and C viruses. Nat Immunol 2019;20(5):613–625. [DOI] [PubMed] [Google Scholar]
- 60.Grifoni A, Weiskopf D, Ramirez SI, et al. Targets of T Cell Responses to SARS-CoV-2 Coronavirus in Humans with COVID-19 Disease and Unexposed Individuals. Cell 2020;181(7):1489–1501 e1415. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 61.Wu C, Zanker D, Valkenburg S, et al. Systematic identification of immunodominant CD8+ T-cell responses to influenza A virus in HLA-A2 individuals. Proc Natl Acad Sci U S A 2011;108(22):9178–9183. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 62.Geginat G, Schenk S, Skoberne M, Goebel W, Hof H. A novel approach of direct ex vivo epitope mapping identifies dominant and subdominant CD4 and CD8 T cell epitopes from Listeria monocytogenes. J Immunol 2001;166(3):1877–1884. [DOI] [PubMed] [Google Scholar]
- 63.Hammond AS, Klein MR, Corrah T, et al. Mycobacterium tuberculosis genome-wide screen exposes multiple CD8 T cell epitopes. Clin Exp Immunol 2005;140(1):109–116. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 64.Ferretti AP, Kula T, Wang Y, et al. Unbiased Screens Show CD8(+) T Cells of COVID-19 Patients Recognize Shared Epitopes in SARS-CoV-2 that Largely Reside outside the Spike Protein. Immunity 2020. [DOI] [PMC free article] [PubMed]
- 65.Swadling L, Diniz MO, Schmidt NM, et al. Pre-existing polymerase-specific T cells expand in abortive seronegative SARS-CoV-2. Nature 2022;601(7891):110–117. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 66.Diniz MO, Mitsi E, Swadling L, et al. Airway-resident T cells from unexposed individuals cross-recognize SARS-CoV-2. Nat Immunol 2022;23(9):1324–1329. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 67.Mateus J, Grifoni A, Tarke A, et al. Selective and cross-reactive SARS-CoV-2 T cell epitopes in unexposed humans. Science 2020;370(6512):89–94. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 68.Schulien I, Kemming J, Oberhardt V, et al. Characterization of pre-existing and induced SARS-CoV-2-specific CD8(+) T cells. Nat Med 2021;27(1):78–85. [DOI] [PubMed] [Google Scholar]
- 69.Quiros-Fernandez I, Poorebrahim M, Fakhr E, Cid-Arregui A. Immunogenic T cell epitopes of SARS-CoV-2 are recognized by circulating memory and naive CD8 T cells of unexposed individuals. EBioMedicine 2021;72:103610. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 70.Nguyen THO, Rowntree LC, Petersen J, et al. CD8(+) T cells specific for an immunodominant SARS-CoV-2 nucleocapsid epitope display high naive precursor frequency and TCR promiscuity. Immunity 2021;54(5):1066–1082 e1065. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 71.Peng Y, Felce SL, Dong D, et al. An immunodominant NP(105–113)-B*07:02 cytotoxic T cell response controls viral replication and is associated with less severe COVID-19 disease. Nat Immunol 2022;23(1):50–61. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 72.Kundu R, Narean JS, Wang L, et al. Cross-reactive memory T cells associate with protection against SARS-CoV-2 infection in COVID-19 contacts. Nat Commun 2022;13(1):80. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 73.Bertagnolio S, Thwin SS, Silva R, et al. Clinical features of, and risk factors for, severe or fatal COVID-19 among people living with HIV admitted to hospital: analysis of data from the WHO Global Clinical Platform of COVID-19. Lancet HIV 2022;9(7):e486–e495. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 74.Liu C, Zhao Y, Okwan-Duodu D, Basho R, Cui X. COVID-19 in cancer patients: risk, clinical features, and management. Cancer Biol Med 2020;17(3):519–527. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 75.Lim S, Bae JH, Kwon HS, Nauck MA. COVID-19 and diabetes mellitus: from pathophysiology to clinical management. Nat Rev Endocrinol 2021;17(1):11–30. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 76.Rydyznski Moderbacher C, Ramirez SI, Dan JM, et al. Antigen-Specific Adaptive Immunity to SARS-CoV-2 in Acute COVID-19 and Associations with Age and Disease Severity. Cell 2020;183(4):996–1012 e1019. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 77.Zhou Y, Chi J, Lv W, Wang Y. Obesity and diabetes as high-risk factors for severe coronavirus disease 2019 (Covid-19). Diabetes Metab Res Rev 2021;37(2):e3377. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 78.Dioverti MV, Razonable RR. Cytomegalovirus. Microbiol Spectr 2016;4(4). [DOI] [PubMed] [Google Scholar]
- 79.Plosa EJ, Esbenshade JC, Fuller MP, Weitkamp JH. Cytomegalovirus infection. Pediatr Rev 2012;33(4):156–163; quiz 163. [DOI] [PubMed] [Google Scholar]
- 80.Bjornevik K, Cortese M, Healy BC, et al. Longitudinal analysis reveals high prevalence of Epstein-Barr virus associated with multiple sclerosis. Science 2022;375(6578):296–301. [DOI] [PubMed] [Google Scholar]
- 81.Lanz TV, Brewer RC, Ho PP, et al. Clonally expanded B cells in multiple sclerosis bind EBV EBNA1 and GlialCAM. Nature 2022;603(7900):321–327. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 82.Ouyang Q, Wagner WM, Wikby A, et al. Large numbers of dysfunctional CD8+ T lymphocytes bearing receptors for a single dominant CMV epitope in the very old. J Clin Immunol 2003;23(4):247–257. [DOI] [PubMed] [Google Scholar]
- 83.Ageing Nikolich-Zugich J. and life-long maintenance of T-cell subsets in the face of latent persistent infections. Nat Rev Immunol 2008;8(7):512–522. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 84.Klenerman P, Oxenius A. T cell responses to cytomegalovirus. Nat Rev Immunol 2016;16(6):367–377. [DOI] [PubMed] [Google Scholar]
- 85.Weber S, Kehl V, Erber J, et al. CMV seropositivity is a potential novel risk factor for severe COVID-19 in non-geriatric patients. PloS one 2022;17(5):e0268530. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 86.Pothast CR, Dijkland RC, Thaler M, et al. SARS-CoV-2-specific CD4(+) and CD8(+) T cell responses can originate from cross-reactive CMV-specific T cells. eLife 2022;11. [DOI] [PMC free article] [PubMed]
- 87.Brenchley JM, Karandikar NJ, Betts MR, et al. Expression of CD57 defines replicative senescence and antigen-induced apoptotic death of CD8+ T cells. Blood 2003;101(7):2711–2720. [DOI] [PubMed] [Google Scholar]
- 88.Jo N, Zhang R, Ueno H, et al. Aging and CMV Infection Affect Pre-existing SARS-CoV-2-Reactive CD8(+) T Cells in Unexposed Individuals. Front Aging 2021;2:719342. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 89.Motozono C, Toyoda M, Zahradnik J, et al. SARS-CoV-2 spike L452R variant evades cellular immunity and increases infectivity. Cell Host Microbe 2021;29(7):1124–1136 e1111. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 90.Grant EJ, Josephs TM, Loh L, et al. Broad CD8(+) T cell cross-recognition of distinct influenza A strains in humans. Nat Commun 2018;9(1):5427. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 91.Krishnamoorthy G, Saxena A, Mars LT, et al. Myelin-specific T cells also recognize neuronal autoantigen in a transgenic mouse model of multiple sclerosis. Nature Medicine 2009;15(6):626–632. [DOI] [PubMed] [Google Scholar]
- 92.Abavisani M, Rahimian K, Mahdavi B, et al. Mutations in SARS-CoV-2 structural proteins: a global analysis. Virol J 2022;19(1):220. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 93.Planas D, Saunders N, Maes P, et al. Considerable escape of SARS-CoV-2 Omicron to antibody neutralization. Nature 2022;602(7898):671–675. [DOI] [PubMed] [Google Scholar]
- 94.Arora P, Kempf A, Nehlmeier I, et al. Omicron sublineage BQ.1.1 resistance to monoclonal antibodies. Lancet Infect Dis 2023;23(1):22–23. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 95.Kurhade C, Zou J, Xia H, et al. Low neutralization of SARS-CoV-2 Omicron BA.2.75.2, BQ.1.1 and XBB.1 by parental mRNA vaccine or a BA.5 bivalent booster. Nat Med 2023;29(2):344–347. [DOI] [PubMed] [Google Scholar]
- 96.Wang Q, Bowen A, Valdez R, et al. Antibody Response to Omicron BA.4-BA.5 Bivalent Booster. N Engl J Med 2023;388(6):567–569. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 97.Jung MK, Jeong SD, Noh JY, et al. BNT162b2-induced memory T cells respond to the Omicron variant with preserved polyfunctionality. Nat Microbiol 2022;7(6):909–917. [DOI] [PubMed] [Google Scholar]
- 98.GeurtsvanKessel CH, Geers D, Schmitz KS, et al. Divergent SARS-CoV-2 Omicron-reactive T and B cell responses in COVID-19 vaccine recipients. Sci Immunol 2022;7(69):eabo2202. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 99.Gao Y, Cai C, Grifoni A, et al. Ancestral SARS-CoV-2-specific T cells cross-recognize the Omicron variant. Nat Med 2022;28(3):472–476. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 100.Brasu N, Elia I, Russo V, et al. Memory CD8(+) T cell diversity and B cell responses correlate with protection against SARS-CoV-2 following mRNA vaccination. Nat Immunol 2022;23(10):1445–1456. [DOI] [PubMed] [Google Scholar]
- 101.Vita R, Mahajan S, Overton JA, et al. The Immune Epitope Database (IEDB): 2018 update. Nucleic Acids Res 2019;47(D1):D339–D343. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 102.Zhang H, Deng S, Ren L, et al. Profiling CD8(+) T cell epitopes of COVID-19 convalescents reveals reduced cellular immune responses to SARS-CoV-2 variants. Cell reports 2021;36(11):109708. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 103.Kim TS, Sun J, Braciale TJ. T cell responses during influenza infection: getting and keeping control. Trends Immunol 2011;32(5):225–231. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 104.Schmidt A, Lapuente D. T Cell Immunity against Influenza: The Long Way from Animal Models Towards a Real-Life Universal Flu Vaccine. Viruses 2021;13(2). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 105.Mansky LM, Temin HM. Lower in vivo mutation rate of human immunodeficiency virus type 1 than that predicted from the fidelity of purified reverse transcriptase. J Virol 1995;69(8):5087–5094. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 106.Mansky LM. Forward mutation rate of human immunodeficiency virus type 1 in a T lymphoid cell line. AIDS Res Hum Retroviruses 1996;12(4):307–314. [DOI] [PubMed] [Google Scholar]
- 107.O’Neil PK, Sun G, Yu H, Ron Y, Dougherty JP, Preston BD. Mutational analysis of HIV-1 long terminal repeats to explore the relative contribution of reverse transcriptase and RNA polymerase II to viral mutagenesis. J Biol Chem 2002;277(41):38053–38061. [DOI] [PubMed] [Google Scholar]
- 108.Abram ME, Ferris AL, Shao W, Alvord WG, Hughes SH. Nature, position, and frequency of mutations made in a single cycle of HIV-1 replication. J Virol 2010;84(19):9864–9878. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 109.Gulzar N, Copeland KF. CD8+ T-cells: function and response to HIV infection. Curr HIV Res 2004;2(1):23–37. [DOI] [PubMed] [Google Scholar]
- 110.Gurdasani D, Iles L, Dillon DG, et al. A systematic review of definitions of extreme phenotypes of HIV control and progression. AIDS 2014;28(2):149–162. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 111.Ramarathinam SH, Gras S, Alcantara S, et al. Identification of Native and Posttranslationally Modified HLA-B*57:01-Restricted HIV Envelope Derived Epitopes Using Immunoproteomics. Proteomics 2018;18(12):e1700253. [DOI] [PubMed] [Google Scholar]
- 112.Migueles SA, Laborico AC, Shupert WL, et al. HIV-specific CD8+ T cell proliferation is coupled to perforin expression and is maintained in nonprogressors. Nat Immunol 2002;3(11):1061–1068. [DOI] [PubMed] [Google Scholar]
- 113.Migueles SA, Osborne CM, Royce C, et al. Lytic granule loading of CD8+ T cells is required for HIV-infected cell elimination associated with immune control. Immunity 2008;29(6):1009–1021. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 114.Migueles SA, Weeks KA, Nou E, et al. Defective human immunodeficiency virus-specific CD8+ T-cell polyfunctionality, proliferation, and cytotoxicity are not restored by antiretroviral therapy. J Virol 2009;83(22):11876–11889. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 115.Edwards BH, Bansal A, Sabbaj S, Bakari J, Mulligan MJ, Goepfert PA. Magnitude of functional CD8+ T-cell responses to the gag protein of human immunodeficiency virus type 1 correlates inversely with viral load in plasma. J Virol 2002;76(5):2298–2305. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 116.Kiepiela P, Ngumbela K, Thobakgale C, et al. CD8+ T-cell responses to different HIV proteins have discordant associations with viral load. Nat Med 2007;13(1):46–53. [DOI] [PubMed] [Google Scholar]
- 117.Barton JP, Goonetilleke N, Butler TC, Walker BD, McMichael AJ, Chakraborty AK. Relative rate and location of intra-host HIV evolution to evade cellular immunity are predictable. Nat Commun 2016;7:11660. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 118.Gaiha GD, Rossin EJ, Urbach J, et al. Structural topology defines protective CD8(+) T cell epitopes in the HIV proteome. Science 2019;364(6439):480–484. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 119.Price DA, Asher TE, Wilson NA, et al. Public clonotype usage identifies protective Gag-specific CD8+ T cell responses in SIV infection. J Exp Med 2009;206(4):923–936. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 120.Chen H, Ndhlovu ZM, Liu D, et al. TCR clonotypes modulate the protective effect of HLA class I molecules in HIV-1 infection. Nat Immunol 2012;13(7):691–700. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 121.Kosmrlj A, Read EL, Qi Y, et al. Effects of thymic selection of the T-cell repertoire on HLA class I-associated control of HIV infection. Nature 2010;465(7296):350–354. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 122.Gorin AM, Du Y, Liu FY, et al. HIV-1 epitopes presented by MHC class I types associated with superior immune containment of viremia have highly constrained fitness landscapes. PLoS pathogens 2017;13(8):e1006541. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 123.Wherry EJ, Kurachi M. Molecular and cellular insights into T cell exhaustion. Nat Rev Immunol 2015;15(8):486–499. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 124.Ndhlovu ZM, Kamya P, Mewalal N, et al. Magnitude and Kinetics of CD8+ T Cell Activation during Hyperacute HIV Infection Impact Viral Set Point. Immunity 2015;43(3):591–604. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 125.Shakiba M, Zumbo P, Espinosa-Carrasco G, et al. TCR signal strength defines distinct mechanisms of T cell dysfunction and cancer evasion. J Exp Med 2022;219(2). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 126.Gate D, Saligrama N, Leventhal O, et al. Clonally expanded CD8 T cells patrol the cerebrospinal fluid in Alzheimer’s disease. Nature 2020;577(7790):399–404. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 127.Dhanwani R, Pham J, Premlal ALR, et al. T Cell Responses to Neural Autoantigens Are Similar in Alzheimer’s Disease Patients and Age-Matched Healthy Controls. Front Neurosci 2020;14:874. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 128.Xu H, Jia J. Single-Cell RNA Sequencing of Peripheral Blood Reveals Immune Cell Signatures in Alzheimer’s Disease. Front Immunol 2021;12:645666. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 129.Merlini M, Kirabali T, Kulic L, Nitsch RM, Ferretti MT. Extravascular CD3+ T Cells in Brains of Alzheimer Disease Patients Correlate with Tau but Not with Amyloid Pathology: An Immunohistochemical Study. Neurodegener Dis 2018;18(1):49–56. [DOI] [PubMed] [Google Scholar]
- 130.Joshi C, Sivaprakasam K, Christley S, et al. CSF-Derived CD4+ T-Cell Diversity Is Reduced in Patients With Alzheimer Clinical Syndrome. Neurol Neuroimmunol Neuroinflamm 2022;9(1). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 131.Sulzer D, Alcalay RN, Garretti F, et al. T cells from patients with Parkinson’s disease recognize α-synuclein peptides. Nature 2017;546(7660):656–661. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 132.Lindestam Arlehamn CS, Dhanwani R, Pham J, et al. alpha-Synuclein-specific T cell reactivity is associated with preclinical and early Parkinson’s disease. Nat Commun 2020;11(1):1875. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 133.Singhania A, Pham J, Dhanwani R, et al. The TCR repertoire of α-synuclein-specific T cells in Parkinson’s disease is surprisingly diverse. Scientific Reports 2021;11(1):302. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 134.Wang P, Yao L, Luo M, et al. Single-cell transcriptome and TCR profiling reveal activated and expanded T cell populations in Parkinson’s disease. Cell Discovery 2021;7(1):52. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 135.Galiano-Landeira J, Torra A, Vila M, Bove J. CD8 T cell nigral infiltration precedes synucleinopathy in early stages of Parkinson’s disease. Brain 2020;143(12):3717–3733. [DOI] [PubMed] [Google Scholar]
- 136.Campisi L, Chizari S, Ho JSY, et al. Clonally expanded CD8 T cells characterize amyotrophic lateral sclerosis-4. Nature 2022;606(7916):945–952. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 137.Yazdani S, Seitz C, Cui C, et al. T cell responses at diagnosis of amyotrophic lateral sclerosis predict disease progression. Nature Communications 2022;13(1):6733. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 138.Varrin-Doyer M, Shetty A, Spencer CM, et al. MOG transmembrane and cytoplasmic domains contain highly stimulatory T-cell epitopes in MS. Neurol Neuroimmunol Neuroinflamm 2014;1(2):e20. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 139.Sabatino JJ, Wilson MR, Calabresi PA, Hauser SL, Schneck JP, Zamvil SS. Anti-CD20 therapy depletes activated myelin-specific CD8. Proc Natl Acad Sci U S A 2019;116(51):25800–25807. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 140.Bronge M, Ruhrmann S, Carvalho-Queiroz C, et al. Myelin oligodendrocyte glycoprotein revisited-sensitive detection of MOG-specific T-cells in multiple sclerosis. J Autoimmun 2019;102:38–49. [DOI] [PubMed] [Google Scholar]
- 141.Bronge M, Högelin KA, Thomas OG, et al. Identification of four novel T cell autoantigens and personal autoreactive profiles in multiple sclerosis. Sci Adv 2022;8(17):eabn1823. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 142.Reagin KL, Funk KE. The role of antiviral CD8(+) T cells in cognitive impairment. Curr Opin Neurobiol 2022;76:102603. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 143.Damiano RF, Guedes BF, de Rocca CC, et al. Cognitive decline following acute viral infections: literature review and projections for post-COVID-19. Eur Arch Psychiatry Clin Neurosci 2022;272(1):139–154. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 144.Bu XL, Yao XQ, Jiao SS, et al. A study on the association between infectious burden and Alzheimer’s disease. Eur J Neurol 2015;22(12):1519–1525. [DOI] [PubMed] [Google Scholar]
- 145.Bordon Y Linking Epstein-Barr virus infection to multiple sclerosis. Nat Rev Immunol 2022;22(3):143. [DOI] [PubMed] [Google Scholar]
- 146.Bar-Or A, Banwell B, Berger JR, Lieberman PM. Guilty by association: Epstein-Barr virus in multiple sclerosis. Nat Med 2022;28(5):904–906. [DOI] [PubMed] [Google Scholar]
- 147.Williams GP, Muskat K, Frazier A, et al. Unaltered T cell responses to common antigens in individuals with Parkinson’s disease. J Neurol Sci 2023;444:120510. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 148.Lindestam Arlehamn CS, Pham J, Alcalay RN, et al. Widespread Tau-Specific CD4 T Cell Reactivity in the General Population. J Immunol 2019;203(1):84–92. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 149.Singhania A, Pham J, Dhanwani R, et al. The TCR repertoire of alpha-synuclein-specific T cells in Parkinson’s disease is surprisingly diverse. Sci Rep 2021;11(1):302. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 150.Evavold BD, Allen PM. Separation of IL-4 production from Th cell proliferation by an altered T cell receptor ligand. Science 1991;252(5010):1308–1310. [DOI] [PubMed] [Google Scholar]
- 151.Dressel A, Chin JL, Sette A, Gausling R, Höllsberg P, Hafler DA. Autoantigen recognition by human CD8 T cell clones: enhanced agonist response induced by altered peptide ligands. The Journal of Immunology 1997;159(10):4943–4951. [PubMed] [Google Scholar]
- 152.Genain CP, Zamvil SS. Specific immunotherapy: one size does not fit all. Nat Med 2000;6(10):1098–1100. [DOI] [PubMed] [Google Scholar]
- 153.Dargahi N, Katsara M, Tselios T, et al. Multiple Sclerosis: Immunopathology and Treatment Update. Brain Sci 2017;7(7). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 154.Kappos L, Comi G, Panitch H, et al. Induction of a non-encephalitogenic type 2 T helper-cell autoimmune response in multiple sclerosis after administration of an altered peptide ligand in a placebo-controlled, randomized phase II trial. The Altered Peptide Ligand in Relapsing MS Study Group. Nat Med 2000;6(10):1176–1182. [DOI] [PubMed] [Google Scholar]
- 155.Bielekova B, Goodwin B, Richert N, et al. Encephalitogenic potential of the myelin basic protein peptide (amino acids 83–99) in multiple sclerosis: results of a phase II clinical trial with an altered peptide ligand. Nat Med 2000;6(10):1167–1175. [DOI] [PubMed] [Google Scholar]
- 156.Barnd DL, Lan MS, Metzgar RS, Finn OJ. Specific, major histocompatibility complex-unrestricted recognition of tumor-associated mucins by human cytotoxic T cells. Proc Natl Acad Sci U S A 1989;86(18):7159–7163. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 157.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]
- 158.Caballero OL, Chen YT. Cancer/testis (CT) antigens: potential targets for immunotherapy. Cancer science 2009;100(11):2014–2021. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 159.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]
- 160.Wang RF, Rosenberg SA. Human tumor antigens recognized by T lymphocytes: implications for cancer therapy. J Leukoc Biol 1996;60(3):296–309. [DOI] [PubMed] [Google Scholar]
- 161.Gilboa E The makings of a tumor rejection antigen. Immunity 1999;11(3):263–270. [DOI] [PubMed] [Google Scholar]
- 162.Heemskerk B, Kvistborg P, Schumacher TN. The cancer antigenome. EMBO J 2013;32(2):194–203. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 163.Schumacher TN, Schreiber RD. Neoantigens in cancer immunotherapy. Science 2015;348(6230):69–74. [DOI] [PubMed] [Google Scholar]
- 164.Schumacher TN, Scheper W, Kvistborg P. Cancer Neoantigens. Annu Rev Immunol 2019;37:173–200. [DOI] [PubMed] [Google Scholar]
- 165.Laumont CM, Perreault C. Exploiting non-canonical translation to identify new targets for T cell-based cancer immunotherapy. Cell Mol Life Sci 2018;75(4):607–621. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 166.Starr TK, Jameson SC, Hogquist KA. Positive and negative selection of T cells. Annu Rev Immunol 2003;21:139–176. [DOI] [PubMed] [Google Scholar]
- 167.Doherty PC, Topham DJ, Tripp RA. Establishment and persistence of virus-specific CD4+ and CD8+ T cell memory. Immunol Rev 1996;150:23–44. [DOI] [PubMed] [Google Scholar]
- 168.Topalian SL, Solomon D, Rosenberg SA. Tumor-specific cytolysis by lymphocytes infiltrating human melanomas. J Immunol 1989;142(10):3714–3725. [PubMed] [Google Scholar]
- 169.Reiner SL, Wang ZE, Hatam F, Scott P, Locksley RM. TH1 and TH2 cell antigen receptors in experimental leishmaniasis. Science 1993;259(5100):1457–1460. [DOI] [PubMed] [Google Scholar]
- 170.McHeyzer-Williams MG, Davis MM. Antigen-specific development of primary and memory T cells in vivo. Science 1995;268(5207):106–111. [DOI] [PubMed] [Google Scholar]
- 171.Walker PR, Ohteki T, Lopez JA, MacDonald HR, Maryanski JL. Distinct phenotypes of antigen-selected CD8 T cells emerge at different stages of an in vivo immune response. J Immunol 1995;155(7):3443–3452. [PubMed] [Google Scholar]
- 172.MacDonald HR, Casanova JL, Maryanski JL, Cerottini JC. Oligoclonal expansion of major histocompatibility complex class I-restricted cytolytic T lymphocytes during a primary immune response in vivo: direct monitoring by flow cytometry and polymerase chain reaction. J Exp Med 1993;177(5):1487–1492. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 173.Altman JD, Moss PA, Goulder PJ, et al. Phenotypic analysis of antigen-specific T lymphocytes. Science 1996;274(5284):94–96. [DOI] [PubMed] [Google Scholar]
- 174.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]
- 175.Molldrem JJ, Lee PP, Wang C, Champlin RE, Davis MM. A PR1-human leukocyte antigen-A2 tetramer can be used to isolate low-frequency cytotoxic T lymphocytes from healthy donors that selectively lyse chronic myelogenous leukemia. Cancer Res 1999;59(11):2675–2681. [PubMed] [Google Scholar]
- 176.Labarriere N, Pandolfino MC, Raingeard D, et al. Frequency and relative fraction of tumor antigen-specific T cells among lymphocytes from melanoma-invaded lymph nodes. Int J Cancer 1998;78(2):209–215. [DOI] [PubMed] [Google Scholar]
- 177.Romero P, Dunbar PR, Valmori D, et al. Ex vivo staining of metastatic lymph nodes by class I major histocompatibility complex tetramers reveals high numbers of antigen-experienced tumor-specific cytolytic T lymphocytes. J Exp Med 1998;188(9):1641–1650. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 178.Lee PP, Yee C, Savage PA, et al. Characterization of circulating T cells specific for tumor-associated antigens in melanoma patients. Nat Med 1999;5(6):677–685. [DOI] [PubMed] [Google Scholar]
- 179.De Plaen E, Lurquin C, Van Pel A, et al. Immunogenic (tum-) variants of mouse tumor P815: cloning of the gene of tum- antigen P91A and identification of the tum- mutation. Proc Natl Acad Sci U S A 1988;85(7):2274–2278. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 180.Wolfel T, Hauer M, Schneider J, et al. A p16INK4a-insensitive CDK4 mutant targeted by cytolytic T lymphocytes in a human melanoma. Science 1995;269(5228):1281–1284. [DOI] [PubMed] [Google Scholar]
- 181.Matsushita H, Vesely MD, Koboldt DC, et al. Cancer exome analysis reveals a T-cell-dependent mechanism of cancer immunoediting. Nature 2012;482(7385):400–404. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 182.Robbins PF, Lu YC, El-Gamil M, et al. Mining exomic sequencing data to identify mutated antigens recognized by adoptively transferred tumor-reactive T cells. Nat Med 2013;19(6):747–752. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 183.van Rooij N, van Buuren MM, Philips D, et al. Tumor exome analysis reveals neoantigen-specific T-cell reactivity in an ipilimumab-responsive melanoma. J Clin Oncol 2013;31(32):e439–442. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 184.Robbins PF, El-Gamil M, Li YF, et al. A mutated beta-catenin gene encodes a melanoma-specific antigen recognized by tumor infiltrating lymphocytes. J Exp Med 1996;183(3):1185–1192. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 185.Lennerz V, Fatho M, Gentilini C, et al. The response of autologous T cells to a human melanoma is dominated by mutated neoantigens. Proc Natl Acad Sci U S A 2005;102(44):16013–16018. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 186.Toebes M, Coccoris M, Bins A, et al. Design and use of conditional MHC class I ligands. Nat Med 2006;12(2):246–251. [DOI] [PubMed] [Google Scholar]
- 187.Rodenko B, Toebes M, Hadrup SR, et al. Generation of peptide-MHC class I complexes through UV-mediated ligand exchange. Nat Protoc 2006;1(3):1120–1132. [DOI] [PubMed] [Google Scholar]
- 188.Newell EW, Klein LO, Yu W, Davis MM. Simultaneous detection of many T-cell specificities using combinatorial tetramer staining. Nat Methods 2009;6(7):497–499. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 189.Hadrup SR, Bakker AH, Shu CJ, et al. Parallel detection of antigen-specific T-cell responses by multidimensional encoding of MHC multimers. Nat Methods 2009;6(7):520–526. [DOI] [PubMed] [Google Scholar]
- 190.Newell EW, Sigal N, Bendall SC, Nolan GP, Davis MM. Cytometry by time-of-flight shows combinatorial cytokine expression and virus-specific cell niches within a continuum of CD8+ T cell phenotypes. Immunity 2012;36(1):142–152. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 191.Newell EW, Sigal N, Nair N, Kidd BA, Greenberg HB, Davis MM. Combinatorial tetramer staining and mass cytometry analysis facilitate T-cell epitope mapping and characterization. Nat Biotechnol 2013;31(7):623–629. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 192.Simoni Y, Becht E, Fehlings M, et al. Bystander CD8(+) T cells are abundant and phenotypically distinct in human tumour infiltrates. Nature 2018;557(7706):575–579. [DOI] [PubMed] [Google Scholar]
- 193.Duhen T, Duhen R, Montler R, et al. Co-expression of CD39 and CD103 identifies tumor-reactive CD8 T cells in human solid tumors. Nat Commun 2018;9(1):2724. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 194.Scheper W, Kelderman S, Fanchi LF, et al. Low and variable tumor reactivity of the intratumoral TCR repertoire in human cancers. Nat Med 2019;25(1):89–94. [DOI] [PubMed] [Google Scholar]
- 195.Rosato PC, Wijeyesinghe S, Stolley JM, et al. Virus-specific memory T cells populate tumors and can be repurposed for tumor immunotherapy. Nat Commun 2019;10(1):567. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 196.Ning J, Gavil NV, Wu S, et al. Functional virus-specific memory T cells survey glioblastoma. Cancer Immunol Immunother 2022;71(8):1863–1875. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 197.Oliveira G, Stromhaug K, Klaeger S, et al. Phenotype, specificity and avidity of antitumour CD8(+) T cells in melanoma. Nature 2021;596(7870):119–125. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 198.Caushi JX, Zhang J, Ji Z, et al. Transcriptional programs of neoantigen-specific TIL in anti-PD-1-treated lung cancers. Nature 2021;596(7870):126–132. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 199.Lowery FJ, Krishna S, Yossef R, et al. Molecular signatures of antitumor neoantigen-reactive T cells from metastatic human cancers. Science 2022;375(6583):877–884. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 200.Puig-Saus C, Sennino B, Peng S, et al. Neoantigen-targeted CD8(+) T cell responses with PD-1 blockade therapy. Nature 2023;615(7953):697–704. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 201.Bentzen AK, Marquard AM, Lyngaa R, et al. Large-scale detection of antigen-specific T cells using peptide-MHC-I multimers labeled with DNA barcodes. Nat Biotech 2016;34(10):1037–1045. [DOI] [PubMed] [Google Scholar]
- 202.Zhang SQ, Ma KY, Schonnesen AA, et al. High-throughput determination of the antigen specificities of T cell receptors in single cells. Nat Biotechnol 2018;36:1156–1159. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 203.Peng S, Zaretsky JM, Ng AHC, et al. Sensitive Detection and Analysis of Neoantigen-Specific T Cell Populations from Tumors and Blood. Cell reports 2019;28(10):2728–2738 e2727. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 204.Lawrence MS, Stojanov P, Polak P, et al. Mutational heterogeneity in cancer and the search for new cancer-associated genes. Nature 2013;499(7457):214–218. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 205.Alexandrov LB, Nik-Zainal S, Wedge DC, et al. Signatures of mutational processes in human cancer. Nature 2013;500(7463):415–421. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 206.Rosenthal R, Cadieux EL, Salgado R, et al. Neoantigen-directed immune escape in lung cancer evolution. Nature 2019;567(7749):479–485. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 207.Zamora AE, Crawford JC, Allen EK, et al. Pediatric patients with acute lymphoblastic leukemia generate abundant and functional neoantigen-specific CD8(+) T cell responses. Sci Transl Med 2019;11(498). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 208.McGranahan N, Furness AJ, Rosenthal R, et al. Clonal neoantigens elicit T cell immunoreactivity and sensitivity to immune checkpoint blockade. Science 2016;351(6280):1463–1469. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 209.Tran E, Ahmadzadeh M, Lu YC, et al. Immunogenicity of somatic mutations in human gastrointestinal cancers. Science 2015;350(6266):1387–1390. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 210.Tran E, Robbins PF, Lu YC, et al. T-Cell Transfer Therapy Targeting Mutant KRAS in Cancer. N Engl J Med 2016;375(23):2255–2262. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 211.Cheng Y, Gunasegaran B, Singh HD, et al. Non-terminally exhausted tumor-resident memory HBV-specific T cell responses correlate with relapse-free survival in hepatocellular carcinoma. Immunity 2021;54(8):1825–1840 e1827. [DOI] [PubMed] [Google Scholar]
- 212.Eberhardt CS, Kissick HT, Patel MR, et al. Functional HPV-specific PD-1(+) stem-like CD8 T cells in head and neck cancer. Nature 2021;597(7875):279–284. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 213.Yau T, Park JW, Finn RS, et al. Nivolumab versus sorafenib in advanced hepatocellular carcinoma (CheckMate 459): a randomised, multicentre, open-label, phase 3 trial. The Lancet Oncology 2022;23(1):77–90. [DOI] [PubMed] [Google Scholar]
- 214.Ferris RL, Blumenschein G Jr., Fayette J, et al. Nivolumab for Recurrent Squamous-Cell Carcinoma of the Head and Neck. N Engl J Med 2016;375(19):1856–1867. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 215.Chow LQM, Haddad R, Gupta S, et al. Antitumor Activity of Pembrolizumab in Biomarker-Unselected Patients With Recurrent and/or Metastatic Head and Neck Squamous Cell Carcinoma: Results From the Phase Ib KEYNOTE-012 Expansion Cohort. J Clin Oncol 2016;34(32):3838–3845. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 216.Cameron BJ, Gerry AB, Dukes J, et al. Identification of a Titin-Derived HLA-A1–Presented Peptide as a Cross-Reactive Target for Engineered MAGE A3–Directed T Cells. Science Translational Medicine 2013;5(197):197ra103–197ra103. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 217.Linette GP, Stadtmauer EA, Maus MV, et al. Cardiovascular toxicity and titin cross-reactivity of affinity-enhanced T cells in myeloma and melanoma. Blood 2013;122(6):863–871. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 218.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]
- 219.Zhu C, McEver RP. Catch bonds: physical models and biological functions. Mol Cell Biomech 2005;2(3):91–104. [PubMed] [Google Scholar]
- 220.Karapetyan AR, Chaipan C, Winkelbach K, et al. TCR Fingerprinting and Off-Target Peptide Identification. Front Immunol 2019;10:2501. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 221.Hudson D, Fernandes RA, Basham M, Ogg G, Koohy H. Can we predict T cell specificity with digital biology and machine learning? Nat Rev Immunol 2023:1–11. [DOI] [PMC free article] [PubMed]
- 222.Schaap-Johansen AL, Vujovic M, Borch A, Hadrup SR, Marcatili P. T Cell Epitope Prediction and Its Application to Immunotherapy. Front Immunol 2021;12:712488. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 223.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]
- 224.Lee CH, Salio M, Napolitani G, Ogg G, Simmons A, Koohy H. Predicting Cross-Reactivity and Antigen Specificity of T Cell Receptors. Front Immunol 2020;11:565096. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 225.Vujovic M, Degn KF, Marin FI, et al. T cell receptor sequence clustering and antigen specificity. Comput Struct Biotechnol J 2020;18:2166–2173. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 226.Katayama Y, Yokota R, Akiyama T, Kobayashi TJ. Machine Learning Approaches to TCR Repertoire Analysis. Front Immunol 2022;13:858057. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 227.Gao Y, Gao Y, Fan Y, et al. Pan-Peptide Meta Learning for T-cell receptor–antigen binding recognition. Nature Machine Intelligence 2023;5(3):236–249. [Google Scholar]
- 228.Wang D, He F, Yu Y, Xu D. Meta-learning for T cell receptor binding specificity and beyond. Nature Machine Intelligence 2023;5(4):337–339. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 229.Bradley P Structure-based prediction of T cell receptor:peptide-MHC interactions. eLife 2023;12. [DOI] [PMC free article] [PubMed]
- 230.Zhang P, Bang S, Cai M, Lee H. Context-Aware Amino Acid Embedding Advances Analysis of TCR-Epitope Interactions. bioRxiv 2023:2023.2004.2012.536635. [Google Scholar]
- 231.Zhou W, Xiang W, Yu J, et al. NeoTCR: an immunoinformatic database of experimentally-supported functional neoantigen-specific TCR sequences. bioRxiv 2023:2023.2002.2013.528383. [Google Scholar]