The molecular interactions underlying regulation of the immune response take place in a nanoscale gap between T cells and antigen-presenting cells, termed the immunological synapse. If these interactions are regulated appropriately, the host is defended against a wide range of pathogens and deranged host cells. If these interactions are disregulated, the host is susceptible to pathogens or tumor escape at one extreme and autoimmunity at the other. Strategies targeting the synapse have helped to establish immunotherapy as a mainstream element in cancer treatment. This Masters' primer will cover the basics of the immunological synapse and some of the applications to tumor immunology. Cancer Immunol Res; 2(11); 1023–33. ©2014 AACR.

No potential conflicts of interest were disclosed.

The following editor(s) reported relevant financial relationships: G. Dranoff — None.

The members of the planning committee have no real or apparent conflicts of interest to disclose.

The T-cell immunological synapse is the junction formed between a T cell and an antigen-presenting cell, comprising interactions for antigen recognition regulating host immunity. If these interactions are dysregulated, the host is susceptible to pathogens or tumor escape at one extreme and autoimmunity at the other. Immunotherapeutic strategies targeting the synapse have shown promise for cancer treatment. Upon completion of this activity, the participant should gain a basic knowledge of the organization and function of the immunological synapse that modulate immune responses.

This activity does not receive commercial support.

T-cell–dependent immune responses protect the host from cancer (1, 2), but also participate in destructive autoimmunity (3, 4). T-cell receptor (TCR) engagement leads to suppression of T-cell locomotion, formation of a specialized junction, and T-cell polarization (5–7). This combination of a specialized junction, cell polarization, and positional stability bears a striking similarity to the classical synapse of the nervous system (8, 9). T cells can also integrate signals through the antigen receptor during migration, and we have referred to this type of mobile, antigen recognition structure as a kinapse (10). The immunological synapse or kinapse integrates three broad categories of receptors: antigen (TCR), adhesion, and costimulatory/checkpoint. The organization of these receptors in the interface affects how they function in a way that cannot be predicted without this spatial and temporal information. The textbook picture of antigen recognition across an approximately 15-nm gap between a T cell and an antigen-presenting cell (APC) provides the justification for why the immune system needs a synapse or kinapse (11). Adhesion molecules, identified first by function with blocking antibodies and then by expression cloning, provide the energy needed to pull cells together, allowing sustained antigen recognition and precise execution of effector functions (12). Finally, costimulatory and checkpoint receptors alter the functional outcome of immunological synapse formation substantially and can also influence the synapse–kinapse balance (13, 14). To date, immunotherapies targeting checkpoint receptors have provided the most promise (15).

Determining how antigen recognition, adhesion, and costimulation/checkpoint receptors come together in the immunological synapse or kinapse required visualization of the close interface between the T cell and the APC. Two major approaches dominated the efforts at molecular analysis. Top-down analysis of T cell–APC pairs using immunofluorescence (16) and bottom-up approaches of applying purified molecules to substrates have both contributed to our understanding of the immunological synapse. Reconstitution methods based on fluid-supported lipid bilayers (SLB), in which the purified molecules are combined in a laterally mobile form, have been effective in recreating the organization of the immunological synapse formed with live T cells (16, 17). The canonical organization of the immune synapse is a bull's eye structure with a central TCR–MHC interaction cluster surrounded by a ring of LFA-1–ICAM-1 adhesion and a distal ring that includes the transmembrane tyrosine phosphatase CD45 (18, 19). Kupfer referred to these radially symmetric compartments as supramolecular activation clusters (SMAC; Fig. 1; ref. 16). The segregation of TCR–MHC interactions from LFA-1–ICAM-1 interactions was a confirmation of the working model formulated by Springer in 1990 that, based on their sizes, the TCR–MHC interactions would need to segregate laterally from the LFA-1–ICAM-1 interactions (12). This is a stereotypical response of helper, cytotoxic, and regulatory T cells (20–23). The SMACs of a stable synapse correspond to polarized compartments in a kinase (24). T cells and dendritic cells (DC) have a more complex organization of similar compartments, perhaps due to the intricate topology of DCs (25, 26). We first consider the three categories of receptors that are organized in the synapse and then how they are coalesced into a functional synapse or kinapse.

Figure 1.

Model immunological synapse—elevated (A) and front-on view (B) of model for synapse focusing on interface. Color code: gray, F-actin lamellipodium (dSMAC); blue, LFA-1–ICAM-1 adhesion (pSMAC); green, CD28–CD80 costimulatory molecules (Endo-cSMAC); and red, TCR–MHC antigen recognition (Exo-cSMAC). The yellow dots are TCR microclusters that include both CD28 (green) and TCR (red). This is where signals are generated.

Figure 1.

Model immunological synapse—elevated (A) and front-on view (B) of model for synapse focusing on interface. Color code: gray, F-actin lamellipodium (dSMAC); blue, LFA-1–ICAM-1 adhesion (pSMAC); green, CD28–CD80 costimulatory molecules (Endo-cSMAC); and red, TCR–MHC antigen recognition (Exo-cSMAC). The yellow dots are TCR microclusters that include both CD28 (green) and TCR (red). This is where signals are generated.

Close modal

T-cell antigen receptor

TCR interaction with MHC–peptide complexes controls the specificity of the immune response and the source of antigens in both cellular and humoral responses. TCR genes undergo rearrangement similar to that of antibody genes (27). T cells with subunits encoded by the α and β genes make up the classical diverse repertoire of naïve T cells that can respond to pathogens and tumors. T cells with subunits encoded by the γ and δ genes include specialized cells that are not MHC restricted. The complete TCR on the cell surface is composed of two highly diversified antigen recognition subunits that are essentially unique for each T cell, either αβ or γδ, in a noncovalent complex with nonpolymorphic CD3ϵ–CD3δ, CD3ϵ–CD3γ, and CD3ζ–CD3ζ dimers (28). The entire complex is critical for surface expression and signal transduction, but it lacks any intrinsic catalytic activity. Instead, the TCR complex has 10 immunotyrosine activation motifs (ITAM) with paired tyrosine residues that, when phosphorylated, can recruit the nonreceptor tyrosine kinase zeta-associated protein of 70 kDa (ZAP-70; Fig. 2; refs. 29, 30).

Figure 2.

SMACs and representative receptor classes in immunological synapse. See text for description and citations.

Figure 2.

SMACs and representative receptor classes in immunological synapse. See text for description and citations.

Close modal

Although it was recognized by 1975 that cell surface–expressed gene products of the highly polymorphic MHC were required to “restrict” T-cell recognition of antigen (31), the full molecular picture of how antigenic peptides bound to MHC was not clear until the crystal structure of an MHC molecule was solved in 1987 (32). There are two distinct types of MHC that control antigen recognition—one set of genes for cytotoxic T cells (CTL) that bind endogenously synthesized cytoplasmic peptides generated by the proteasome (class I), and a distinct set of genes for helper T (Th) cells that bind exogenous antigens taken into the cell by endocytosis (class II). The process of cross-presentation or cross-dressing allows specialized APCs needed for T-cell priming to present exogenous antigens through the class I pathway, which is likely critical for anticancer cellular immunity (33). MHC class II molecules can also engage in signaling (Fig. 2; ref. 34). Other MHC-like molecules present nonpeptide antigenic structures. These include CD1d presentation of glycolipids to invariant natural killer (NK) T cells and MR1 presentation of bacterial metabolites to mucosa-associated invariant T cells (MAIT). These innate-like T cells may have utility in immune response to tumors (35).

Conventional αβ T cells are marked by different coreceptors, CD8 and CD4 (36). CD8 binds to nonpolymorphic elements of MHC class I, and CD4 to that of MHC class II. In T-cell development, thymocytes rearrange TCR genes and test receptors for interaction with self-peptides on MHC. Strong interactions lead to apoptosis or, for a limited number of clones, the generation of actively suppressive thymic regulatory T cells (Treg), which are important in tumor immunology as their formation of immunological synapses can suppress antitumor responses in part by destabilizing T-effector immunological synapses (37–40). Differences in signaling between conventional αβ T cells and Tregs offer therapeutic opportunities to push the balance toward regulation in autoimmunity and toward responsiveness in cancer therapy (23, 41). A weaker recognition process is required for thymocyte survival, and this is thought to bias the TCR repertoire toward recognition of MHC with either CD4 or CD8 (42). This weak self-recognition is thought to set the stage for foreign peptide recognition by providing the T cell with weak recognition of the peptide flanking, polymorphic α-helices of the MHC proteins, and also provides survival signals to mature T cells. It is still debated whether the germline-encoded TCRs are evolutionarily biased to recognize MHC molecules in a certain preferred orientation, or if the germline TCR repertoire has an antibody-like lack of bias and entirely learns to recognize MHC molecules in the thymus (43, 44).

In a tumor context, antigen recognition can enhance or inhibit responses. The classical tumor-infiltrating lymphocyte (TIL) is a CD8 CTL that can directly kill tumor cells, and the infiltration of tumors by CD8 T cells correlates with better outcome for some tumors (2). APCs in tumors can include CD11c+ cells with characteristics of DCs (45). Cancer vaccines can promote expansion and differentiation of T cells with antitumor specificity in a therapeutic setting (46). T-cell specificity can also be engineered through introduction of chimeric antigen receptors into patient cells in adoptive immunotherapy (47).

Adhesion receptors

Adhesion molecules are critical for sensitive antigen recognition required for tumor rejection (48). Strategies based on immunizing mice with CTL lines followed by selection for mAbs that block killing led to the discovery of lymphocyte function–associated antigen-1 (now CD11a/CD18, but often referred to as LFA-1), LFA-2 (now CD2), and LFA-3 (now CD58; Fig. 2; ref. 49).

CD2 and CD58 define a heterophilic adhesion receptor pair (50). CD2 and CD58 are members of the immunoglobulin (Ig) superfamily and are similar in size to the TCR and MHC–peptide complex, suggesting that CD2 may cooperate very closely with the TCR (12, 51). CD2 and CD58 are members of the signaling lymphocyte activation molecule (SLAM) family. Most other SLAM family members are homophilic (they mediate adhesion by binding between products of the same gene) that interact through their cytoplasmic domain with SLAM-associated protein (SAP), which links to the tyrosine kinase Fyn (52). SLAM family members are required for particular interactions between thymocytes in the development of certain specialized T-cell lineages and for early T-cell–B-cell interactions required for germinal center formation (53).

LFA-1 is a heterodimer with a unique α subunit that shares a β subunit with three other cell-surface heterodimers, each of which has an α subunit with a distinct expression pattern. Functional screening for mAbs that block LFA-1–dependent aggregation after immunization with B cells from LFA-1–deficient patients identified the first LFA-1 ligand, intercellular adhesion molecule-1 (ICAM-1 or CD54; ref. 54). LFA-1 is a member of the integrin family, and ICAM-1 is a member of the Ig superfamily. The intercellular link formed by the interaction of LFA-1 with ICAM-1 is much longer than that with the TCR, and it was suggested that the TCR–MHC and LFA-1–ICAM-1 interactions would need to segregate laterally in the interface, the first prediction on the structure of the immunological synapse (12). ICAM-2, a second ligand for LFA-1 that was cloned from an endothelial cell library, is also an Ig superfamily member (55). ICAM-3 was defined by functional blocking in a screen in which ICAM-1 and ICAM-2 were blocked (56). T cells express other adhesion molecules including additional integrins (57). Integrin ligands are expressed on all nucleated cells and can be further induced, in the case of ICAM-1, by exposure of stromal cells to inflammatory cytokines, including TNF, IL1β, or IFNγ (58). A complication of the therapeutic use of anti–LFA-1 mAbs is that LFA-1 is important for interactions with endothelial cells (59), which are important for optimal localization of cells, in addition to immunological synapse formation. Anti–LFA-1 mAbs were approved for treatment of psoriasis, but the therapeutic window between reduction of disease and increased vulnerability to infection was too small (60). There are activating Abs to LFA-1, but the affinity of LFA-1 for ICAMs is optimized to allow rapid leukocyte migration, and constitutively active LFA-1 is defective in mediating migration, contraindicating LFA-1 affinity enhancement as a therapeutic strategy (61).

Although adhesion molecules are diverse in structure, both integrins and the SLAM family signal through the Src family kinase Fyn rather than Lck, which is the primary partner for antigen and costimulatory receptors (62). Another common feature is that both LFA-1 and CD2 engage ligands in the periphery of the immunological synapse, even though the CD2–CD58 interactions are the correct size to colocalize with TCR in the center (Fig. 2; see Supplementary Information in ref. 17).

Costimulation

Costimulatory receptors have minimal signaling or adhesive activity on their own, but can enhance adhesion and signaling locally when combined with other stimuli, primarily through the TCR (63, 64). Costimulatory receptors were predicted as a corollary of the clonal selection model, in which T cells must be able to attenuate their responses to harmless foreign proteins from the environment that are not present in the thymus and could drive inappropriate immune responses if not checked by some mechanism for extrathymic tolerance (65). Pathogens or tumors would need to upregulate costimulation to drive a T cell to respond, and incorporation of costimulatory ligands directly into tumor cells has been implemented in tumor vaccines (66). Activation of mature T cells in the absence of costimulation leads to a second round of clonal deletion that protects the host against immune responses to harmless environmental antigens. Jenkins and Schwartz (67) discovered that antigen recognition with suboptimal costimulation could induce a state of nonresponsiveness that they named anergy.

The archetypal costimulator is CD28, which is an Ig superfamily member with a homodimeric structure and a cytoplasmic domain lacking enzymatic activity but containing motifs that recruit and activate Lck and indirectly, protein kinase C (PKC)-θ, an important PKC isoform in T cells that contributes to the activation of NF-κB transcription factors and promotes IL2 production (Fig. 2; ref. 68). The activity of CD28 is dependent upon the upregulation of B7-1 (CD80) and B7-2 (CD86) on APCs and the interaction in an immunological synapse (26). Both CD80 and CD86 are upregulated on DCs by stimulation through maturation signals delivered by Toll-like receptors and inflammatory cytokines (69, 70). This is a critical link between innate and adaptive immunity, and the potential of some tumors to grow without inducing expression of costimulatory ligands may allow them to tolerize tumor-specific T cells.

The inducible T-cell costimulator (ICOS, CD278) is a second Ig superfamily costimulatory system. In contrast with CD28, it is not expressed on naïve T cells, but is upregulated following T-cell activation and binds ligand of ICOS (LICOS, CD275; ref. 71). LICOS is expressed prominently on DCs and B cells, but can be induced on stromal cells by inflammatory cytokines. ICOS is critical for germinal center reactions, in which its ability to activate phosphatidylinositol-3 kinase is particularly important (72). In the context of cancer immunotherapy, ICOS costimulation promotes Th17 differentiation in CD4 T cells, which is an effective response mode for some solid tumors (71).

The other large costimulatory receptors are members of the TNF receptor (TNFR) superfamily, including CD27, GITR (CD357), 4-1BB (CD137), and OX40 (CD134). These receptors signal through TNFR-associated factors (TRAF) using K63 ubiquitination to signal through activation of NF-κB2 transcription factors (73, 74). With the exception of CD27, which is constitutively expressed, these costimulatory receptors are induced on conventional T cells in hours to days following initial activation. Therefore, they are involved in effector and memory functions of T cells following the initial expansion phase. These receptors are also constitutively expressed on Tregs, and this perhaps offers an opportunity to modulate negative regulation in the tumor microenvironment (TME). The last set of costimulatory receptors are members of the lectin-like receptor superfamily that pair noncovalently with DAP10, a disulfide-linked heterodimer containing a sequence motif that allows recruitment of phosphatidylinositol-3-kinase, which contributes to activation of the growth-promoting kinase AKT (75). These receptors, with NKG2D as a prime example, are expressed on CTLs as well as NK cells and promote killing. The counter-receptors for NKG2D are induced by cellular stress and are potentially important in recognition of deranged host cells (69, 76). However, tumor cells proteolytically shed one of the human counter-receptors, MICA, leading to jamming of this system as an escape mechanism (77).

There is evidence for combinations of adhesion and costimulatory ligands generating partial signals that can lead to transient formation of ligand-independent immunological synapses. This has been observed with NKG2D in human (22) and mouse (78) CTLs and recently for CD28 in Th cells (79). In this context, the upregulation of ICAM-1, CD80, and NKG2D ligands such as MICA (human) and Rae-1 (mice) can induce a state of heightened surveillance in inflamed or stressed tissues.

Checkpoint blockade

Coinhibitory or checkpoint receptors are a natural complement to the costimulators—they use various negative signaling pathways, often recruiting tyrosine phosphatases, such as SHP-1 and SHP-2, to attenuate tyrosine kinase cascades or other signaling pathways.

One of the best-defined checkpoint inhibitors is cytotoxic T-lymphocyte antigen-4 (CTLA-4, CD152), which is induced rapidly following T-cell activation (80, 81). CTLA-4 has a constitutively active internalization motif in its cytoplasmic domain such that it does not accumulate on the cell surface except when ligated in the synapse or when the Y-based internalization motif is phosphorylated (82). CTLA-4 competes with CD28 for binding to CD80 more potently than for CD86 (83). In Tregs, CTLA-4 is constitutively expressed, and coupling to the endocytic pathway leads to the removal of CD80 and CD86 from APCs, which impairs CD28-dependent responses of other T cells (84). CTLA-4 associates with PKCϵ in Tregs (Fig. 2; ref. 85). Anti–CTLA-4 mAbs are approved for treatment of melanoma (86). Although CTLA-4–deficient mice suffer from fatal immunopathology (87), there is a significant therapeutic window in which antitumor effects can be achieved with limited toxicity.

Another checkpoint receptor is programmed cell death-1 (PD-1, CD279). PD-1 binds PD-1 ligands 1 and 2 (PD-L1, CD274 and PD-L2, CD273; refs. 88, 89) and is recruited to the immunological synapse in a manner related to MHC–peptide strength and abundance (90, 91). PD-1 recruits SHP-2 tyrosine phosphatase and attenuates early TCR signaling upon ligand binding (Fig. 2; ref. 92). The impact of PD-1 on the stability of the immunological synapse depends upon the context, with destabilization in autoimmune and delayed-type hypersensitivity (93, 94), but stabilization in antiviral responses (95). Stabilization of the immunological synapse in this context results in an immune paralysis in which the T cell cannot respond nor be released to engage other targets. PD-1 is associated with T-cell exhaustion in chronic viral infection, and this mechanism can protect the host from immunopathology related to toxicity of chronic immune responses (96). However, it can be exploited by pathogens and tumors as an evasion mechanism (97). Recently, mAbs to PD-1 have been approved for treatment of melanoma in Japan and the United States; so far, results in melanoma and lung cancer are promising (98). The ability to predict patient response based on information about the tumor is a significant goal of personalized medicine. Trials are in progress to determine if PD-L1 expression on the tumor predicts patient response. PD-1 is expressed on Tregs and suppresses its activity. Thus, blockade of PD-1 may increase Treg function, suggesting a rationale for combining anti–PD-1 with anti–CTLA-4 (99), as the latter suppresses Treg function. Other checkpoint inhibitors include Vista and SIRPα (CD172A; refs. 100, 101).

Hierarchy of interactions

Antigen, adhesion, and costimulatory/checkpoint receptors are highly interdependent. LFA-1 activity on T cells is increased by TCR signaling in a process referred to as inside-out signaling (102). This observation leads to a “chicken and egg” problem for initiating TCR signaling in which LFA-1 clearly is important for T-cell sensitivity to antigen (103), yet TCR signaling is necessary for activation of LFA-1. A likely solution involves chemokines in the tissue microenvironment, which stimulate the activity of LFA-1 at the leading edge of T cells and establish a low level of transient LFA-1–dependent adhesion, thus increasing the chances that TCR can detect rare MHC–peptide complexes (104). Once the TCR is triggered, this can further enhance LFA-1 activity and promote formation of an immunologic synapse (105). Increases in LFA-1 affinity are driven by large conformational changes in the heterodimers (Fig. 2; ref. 106). Costimulatory receptor CD28 interacts with ligands in a TCR-dependent fashion, with the critical regulatory step being its low expression on quiescent cells (64), indicating that CD28 only engages its ligand at sites “seeded” by TCR microclusters (MC; ref. 13). One way to consider this is that in the steady state, LFA-1–ICAM-1 interactions allow rapid scanning of APC surfaces for TCR–MHC complexes, whose signaling increases LFA-1–dependent adhesion and generates local MCs, which favors the participation of CD2, CD28, and other small adhesion molecules. Some adhesion molecules and costimulatory receptors cooperate in a process called superagonism. Antibody combinations to CD2 monomer (107) or single antibodies to CD28 homodimer (108) can induce full T-cell activation in a TCR-dependent manner. The structural basis of these effects is not known, but it appears that in some extreme conditions induced by antibodies, the normal process of TCR induction of adhesion/costimulation can operate backwards. It is not clear if this happens physiologically, but the phenomenon may be useful in a clinical setting if it can be controlled and directed (109).

TCR microclusters

When TCRs are ligated by MHC–peptide complexes presented on a cell, e.g., anti-CD3 antibodies on a solid substrate or MHC–peptide complexes and ICAM-1 on a supported planar bilayer, the first structures that can be visualized at the interface are sub-micron TCR clusters that form in an F-actin–dependent manner (110–113). It has been argued that TCRs are preclustered on T cells, but the ligand-induced clusters have a number of distinct characteristics (79, 114). Ligand-induced MCs recruit the src family kinase Lck to the TCR, leading to the phosphorylation of ITAMs and the recruitment and activation of ZAP-70. ZAP-70 phosphorylates the membrane-anchored linker of activated T cells (LAT) that recruits phospholipase Cγ (PLCγ) and facilitates its activation by the inducible T-cell kinase (ITK; ref. 115). PLCγ-mediated generation of diacylglycerol and inositol-1,3,5-trisphosphate leads to the activation of Ras, PKC, and calcium ion effector pathways that can account for transcriptional activation of the T-cell growth and regulatory cytokines such as IL2 (116, 117). TCR can use a module, including the guanine nucleotide exchange factor Vav that activates Notch, to directly control Myc transcription and T-cell proliferation independent of IL2 (118).

Src family kinases Fyn (mentioned in adhesion context) and Lck (dominant kinase downstream of TCR) are regulated by intramolecular interactions. All Src family kinases have a C-terminal tyrosine (Y505 in Lck) that serves as an intramolecular ligand for its own Src homology 2 (SH2) domain. When the SH2 domain is bound to the C-terminal tyrosine in parallel with a second intramolecular interaction of the Src homology 3 (SH3) domain with the polyproline motif between the kinase domain and the SH2 domain, the kinase is locked in an inhibited state. The C-terminal Src kinase (Csk) mediates this phosphorylation when recruited to membrane-anchored Src substrates in the vicinity of the active Src kinase. This negative feedback is counteracted by the transmembrane tyrosine phosphatase CD45. Selective Csk inhibition results in ligand-independent TCR signaling that synergizes with CD28 signals (119). CD45 deficiency results in inhibition of TCR signaling (120). However, CD45 also can reverse activating phosphorylations mediated by Lck and ZAP-70 (121). Interestingly, CD45 is excluded from TCR MCs, and this reduction in CD45 density, based on the large size of its extracellular domain, appears to be important in setting the appropriate local level of phosphatase activity for TCR triggering (113, 122, 123; Fig. 2). Any small adhesion system will generate interactions that exclude CD45, so it is unlikely the CD45 exclusion is sufficient for full TCR signaling, but some level of exclusion appears to be necessary (79, 124).

Supramolecular activation clusters

SMACs defined by Kupfer are divided into the central, peripheral, and distal regions, or cSMAC, pSMAC, and dSMAC (Fig. 1). On the basis of analysis of immunological synapse dynamics using the SLB system, dSMAC behaves like the leading lamellipodium of migrating cells in that it undergoes cycles of protrusion and retraction (24). In contrast, pSMAC is rich in LFA-1 and the integrin to cytoskeletal linker talin, defining it as a lamellum, based on an abrupt change in F-actin dynamics and filament decorations compared with the lamellipodium (24, 125). Higher-resolution imaging has forced a revision of Kupfer's SMAC definitions. First, the active TCR–MHC interactions that lead to signaling in the immunological synapse are concentrated in small MCs that are formed in the dSMAC and move centripetally through the pSMAC to the cSMAC (111, 113, 126). This traversal of the integrin-rich pSMAC by the smaller TCR is possible because the LFA-1–ICAM-1 interaction also begins in MCs in the dSMAC and consolidates into a reticular network with submicron holes in which the TCR–MHC MCs can be fully segregated (127). The LFA-1–ICAM-1 reticulum in the pSMAC is continually moving inward, which allows the central transport of the TCR–MHC MCs (128). Although the immunological synapse can be stable for hours, the dSMAC and pSMAC are highly dynamic structures that are completely renewed every few minutes (24). The second major revision is the structure of the cSMAC. The original cSMAC, as defined by Kupfer, is a TCR–MHC-rich central structure with accumulation of PKC-θ. We now know that the prominent accumulation of PKC-θ is a signature of CD28-dependent costimulation (68). Work with SLB containing only ICAM-1 and activating MHC–peptide complexes revealed a very different cSMAC in which TCR–MHC interactions were highly stabilized and signaling was terminated (17, 113). Further study of bilayers containing ICAM-1, MHC, and CD80 revealed that CD28 and PKC-θ are initially colocalized with TCR in MCs, but on convergence in the cSMAC segregate into a TCR-rich, CD28-deficient zone, in which signaling is terminated, and a TCR-poor, CD28-rich region, in which CD28-PKC-θ–enhanced TCR signaling continues (13, 26). Further analysis of the TCR–MHC-rich signal termination zone in the cSMAC revealed that it is composed of TCR-enriched extracellular microvesicles that bud from the plasma membrane and become trapped in a persistent interaction with the MHC in the SLB (129). Vesicles that bud from the plasma membrane are defined as ectosomes. The budding process is dependent on tumor-susceptibility gene 101 (TSG101), and the buds are arrested by the expression of dominant-negative vacuolar protein sorting 4 (VPS4), implicating the endosomal sorting complexes required for transport (ESCRT) pathway (Fig. 2; ref. 129). This is a property shared with exosomes. In T-cell–B-cell interactions, these TCR-enriched extracellular vesicles are collected by the B cell and moved away from the interface. CD28–CD80 interactions are efficiently sorted away from the bulk of the TCR that is sorted to the extracellular vesicles. Thus, the cSMAC can be divided into two components: the endo-cSMAC, in which TCR and CD28 continue to signal within the synapse, and the exo-cSMAC, composed of TCR-enriched extracellular vesicles. We have found that the checkpoint receptor PD-1 is highly enriched in the exo-cSMAC. This would suggest that TCR and PD-1 are closely associated and co-sorted or that PD-1 is ubiquitinated. The TCR-enriched vesicles continue to activate signaling in the B cell through engaging surface MHC molecules, and in the case of MHC class II may be relevant to T-cell help delivery as an active form of exo-TCR (129, 130). Studies using the SLB system suggest that all T cells engage in this type of vesicle transfer, opening the possibility that helper, cytotoxic, and regulatory cells may generate exo-TCR in a carrier that can contain other information, including nucleic acids (131). Whether exo-TCR plays a role in the TME is an open question. The modular structure of the immunological synapse thus incorporates a lamellipodium (dSMAC), lamellum (pSMAC), TCR MCs, a sorting domain that can enrich CD28-enhanced TCR signaling complexes (endo-cSMAC), and an extracellular compartment of TCR-enriched extracellular vesicles (exo-cSMAC; Figs. 1 and 2). The exo-cSMAC is a transient compartment that is normally internalized by the APC. When projected into the more complex three-dimensional setting of a T-cell–DC interface, the lamellipodium, lamellum, and multiple endo-cSMACs remain clearly visible at the interface. It is likely that the multifocal close contacts observed in electron microscopy are not MCs, but are multiple, larger endo-cSMAC structures. It has also been proposed that the MCs can manifest as small projections that indent the APC surface, rather than moving laterally (132). There is also an LFA-1–dependent actin cloud or lamellum that imparts a more three-dimensional aspect to the actin cytoskeleton, and this structure incorporates a subset of signaling components (133, 134). This raises issues of subsynaptic vesicular components that may also play an important role in signaling (135, 136).

A central element of the immunological synapse concept is directed secretion of soluble components into the synaptic cleft (137–139). This is fundamental to models of T-cell help and cell-mediated killing of infected cells or tumor cells. A major assumption has been that orientation of the centrioles and Golgi toward the APC reflects directed secretion. This has been challenged by studies using antibody-mediated capture to determine the orientation of secretion (140). In this case, molecules that move through the highly oriented Golgi can be secreted randomly based on the manner in which secretory vesicles are transported after leaving the Golgi apparatus. For example, IFNγ was found to undergo directed secretion from helper T cells, but TNF and chemokines were not directed to the synapse. The TCR can be directly delivered to the synapse in vesicles (141). Cytolytic granules are directly delivered to a secretory domain near the endo-cSMAC compartment (20). Recently, it has been proposed that the centrosome docking at the immunological synapse sets up a de facto primary cilium that enables intraflagellar transport machinery and hedgehog pathway signaling to be implemented in the immunological synapse (142, 143). Despite this high level of specialization, it has been shown that CTLs may operate in a “quick draw” mode and release cytolytic granules at targets without reorienting and docking the centrioles to the synapse (144). NK cells also use more dynamic kinapse in signal integration with tumors, with advantages in the ability to penetrate into the tumor over more stable synaptic interactions (145). However, even for NK cells, converting kinapses into synapses with tumor-targeting antibodies increases the efficiency of killing (146). The potential advantage of synapses is the ability to concentrate the cytolytic components on the target surface. Comparison of synaptic versus kinaptic killing in the context of CD8 versus CD4 cytotoxic T cells in vitro suggests that the advantage of forming a stable synapse is about 3-fold (147, 148). In a murine breast cancer model, anti–CTLA-4 mAbs stimulate motility of activated T cells in the tumor (149), consistent with other evidence that CTLA-4 engagement could stimulate T-cell motility (150). Stabilization of activated T-cell interactions through induction of Rae-1, which binds NKG2D on T cells, was required to decrease tumor growth (149). Antigen recognition processes that lead to stable synapses may be desirable in immunotherapy settings. It will be of great interest to determine the mode of interaction induced by chimeric receptors that combine ITAM and costimulatory signaling in one polypeptide (47), particularly as conventional synapses with B chronic lymphocytic leukemia cells are defective (3). Exocytic trafficking in the synapse is likely to be balanced by endocytic trafficking. One manifestation of endocytic processes is trogocytosis, in which one cell “gnaws” off pieces of another cell. In addition to ESCRT-dependent budding to generate TCR-enriched extracellular vesicles, some fraction of TCRs in the cSMAC regions undergoes internalization and can bring MHC from the APC with it (129, 151). CD8 T cells that capture MHC-peptide from target cells through trogocytosis actually become targets for killing by other CTLs in the culture (152). What controls the ratio of TCR that is transferred to the APCs in extracellular vesicles versus MHC that is captured by the T cell from the APCs is not known, but both processes are readily detectable across a single immunological synapse (129). The consequence of exo-TCR transfer for tumor cells is not known. CTLs have been shown to release TCR-positive vesicles at target cells (153, 154). It is not clear if these are similar to the exo-TCR reported by Choudhuri and colleagues (129) as it is not clear if these exosome-like structures contain recently engaged TCRs or were sorted into these structures without prior ligand engagement through a distinct mechanism of TCR sorting. It may be possible to isolate the vesicles budded from the plasma membrane versus those released from granules and subject these to proteomic analysis and functional testing.

The immunological synapse mode of stable engagement of targets benefits the attack on tumor. One of the challenges is that bona fide tumor antigens are likely to be weaker agonists that may be more dependent on costimulation for stabilization of synapses. Current therapies that target checkpoint receptors are directly manipulating the immunological synapse. Anti–CTLA-4 mAbs may both disinhibit conventional cytotoxic and helper T cells while taking away one mechanism by which Tregs function to reduce suppression in the draining lymphoid tissues, among surrounding DCs and in the TME. The impact of PD-1 blockade on T-cell dynamics is determined by the context, including integration of other costimulatory signals. Potential autoimmune toxicity of anti–PD-1 may be offset by the enhancement of Treg function due to blockade of PD-1 on Tregs. Thus, anti–CTLA-4 and anti–PD-1 may be complementary in the TME by offsetting any enhancement of Treg function from PD-1 with blockade by anti–CTLA-4 (99). Antibodies can also have agonistic effects, particularly if Fc binding is intact, leading to additional parameters. TCR-enriched extracellular vesicles, unique structures that share properties with both ectosomes and exosomes, may also have multiple roles in the TME. Further study of the immunological synapse in the TME is needed to appreciate other interactions within and between SMACs to achieve optimal results for immunotherapy.

The author thanks members of the Dustin laboratory and collaborators for stimulating discussions and many insights.

M.L. Dustin was supported by NIH grants AI043542 and AI55037, a Principal Research Fellowship from the Wellcome Trust, and a Senior Research Fellowship from the Kennedy Trust for Rheumatology.

1.
Shankaran
V
,
Ikeda
H
,
Bruce
AT
,
White
JM
,
Swanson
PE
,
Old
LJ
, et al
IFNgamma and lymphocytes prevent primary tumour development and shape tumour immunogenicity
.
Nature
2001
;
410
:
1107
11
.
2.
Galon
J
,
Costes
A
,
Sanchez-Cabo
F
,
Kirilovsky
A
,
Mlecnik
B
,
Lagorce-Pages
C
, et al
Type, density, and location of immune cells within human colorectal tumors predict clinical outcome
.
Science
2006
;
313
:
1960
4
.
3.
Ramsay
AG
,
Johnson
AJ
,
Lee
AM
,
Gorgun
G
,
Le Dieu
R
,
Blum
W
, et al
Chronic lymphocytic leukemia T cells show impaired immunological synapse formation that can be reversed with an immunomodulating drug
.
J Clin Invest
2008
;
118
:
2427
37
.
4.
Schubert
DA
,
Gordo
S
,
Sabatino
JJ
 Jr
,
Vardhana
S
,
Gagnon
E
,
Sethi
DK
, et al
Self-reactive human CD4 T cell clones form unusual immunological synapses
.
J Exp Med
2012
;
209
:
335
52
.
5.
Kupfer
A
,
Singer
SJ
. 
The specific interaction of helper T cells and antigen-presenting B cells. IV. Membrane and cytoskeletal reorganizations in the bound T cell as a function of antigen dose
.
J Exp Med
1989
;
170
:
1697
713
.
6.
Dustin
ML
,
Bromley
SK
,
Kan
Z
,
Peterson
DA
,
Unanue
ER
. 
Antigen receptor engagement delivers a stop signal to migrating T lymphocytes
.
Proc Natl Acad Sci U S A
1997
;
94
:
3909
13
.
7.
Mempel
TR
,
Henrickson
SE
,
Von Andrian
UH
. 
T-cell priming by dendritic cells in lymph nodes occurs in three distinct phases
.
Nature
2004
;
427
:
154
9
.
8.
Dustin
ML
,
Olszowy
MW
,
Holdorf
AD
,
Li
J
,
Bromley
S
,
Desai
N
, et al
A novel adapter protein orchestrates receptor patterning and cytoskeletal polarity in T cell contacts
.
Cell
1998
;
94
:
667
77
.
9.
Dustin
ML
,
Colman
DR
. 
Neural and immunological synaptic relations
.
Science
2002
;
298
:
785
9
.
10.
Dustin
ML
. 
Cell adhesion molecules and actin cytoskeleton at immune synapses and kinapses
.
Curr Opin Cell Biol
2007
;
19
:
529
33
.
11.
Garcia
KC
,
Degano
M
,
Stanfield
RL
,
Brunmark
A
,
Jackson
MR
,
Peterson
PA
, et al
An alphabeta T cell receptor structure at 2.5 A and its orientation in the TCR-MHC complex
.
Science
1996
;
274
:
209
19
.
12.
Springer
TA
. 
Adhesion receptors of the immune system
.
Nature
1990
;
346
:
425
34
.
13.
Yokosuka
T
,
Kobayashi
W
,
Sakata-Sogawa
K
,
Takamatsu
M
,
Hashimoto-Tane
A
,
Dustin
ML
, et al
Spatiotemporal regulation of T cell costimulation by TCR-CD28 microclusters and protein kinase C theta translocation
.
Immunity
2008
;
29
:
589
601
.
14.
Korman
AJ
,
Peggs
KS
,
Allison
JP
. 
Checkpoint blockade in cancer immunotherapy
.
Adv Immunol
2006
;
90
:
297
339
.
15.
Callahan
MK
,
Wolchok
JD
. 
At the bedside: CTLA-4- and PD-1-blocking antibodies in cancer immunotherapy
.
J Leukoc Biol
2013
;
94
:
41
53
.
16.
Monks
CR
,
Freiberg
BA
,
Kupfer
H
,
Sciaky
N
,
Kupfer
A
. 
Three-dimensional segregation of supramolecular activation clusters in T cells
.
Nature
1998
;
395
:
82
6
.
17.
Grakoui
A
,
Bromley
SK
,
Sumen
C
,
Davis
MM
,
Shaw
AS
,
Allen
PM
, et al
The immunological synapse: a molecular machine controlling T cell activation
.
Science
1999
;
285
:
221
7
.
18.
Johnson
KG
,
Bromley
SK
,
Dustin
ML
,
Thomas
ML
. 
A supramolecular basis for CD45 tyrosine phosphatase regulation in sustained T cell activation
.
Proc Natl Acad Sci U S A
2000
;
97
:
10138
43
.
19.
Freiberg
BA
,
Kupfer
H
,
Maslanik
W
,
Delli
J
,
Kappler
J
,
Zaller
DM
, et al
Staging and resetting T cell activation in SMACs
.
Nat Immunol
2002
;
3
:
911
7
.
20.
Stinchcombe
JC
,
Bossi
G
,
Booth
S
,
Griffiths
GM
. 
The immunological synapse of CTL contains a secretory domain and membrane bridges
.
Immunity
2001
;
15
:
751
61
.
21.
Potter
TA
,
Grebe
K
,
Freiberg
B
,
Kupfer
A
. 
Formation of supramolecular activation clusters on fresh ex vivo CD8+ T cells after engagement of the T cell antigen receptor and CD8 by antigen-presenting cells
.
Proc Natl Acad Sci U S A
2001
;
98
:
12624
9
.
22.
Somersalo
K
,
Anikeeva
N
,
Sims
TN
,
Thomas
VK
,
Strong
RK
,
Spies
T
, et al
Cytotoxic T lymphocytes form an antigen-independent ring junction
.
J Clin Invest
2004
;
113
:
49
57
.
23.
Zanin-Zhorov
A
,
Ding
Y
,
Kumari
S
,
Attur
M
,
Hippen
KL
,
Brown
M
, et al
Protein kinase C-theta mediates negative feedback on regulatory T cell function
.
Science
2010
;
328
:
372
6
.
24.
Sims
TN
,
Soos
TJ
,
Xenias
HS
,
Dubin-Thaler
B
,
Hofman
JM
,
Waite
JC
, et al
Opposing effects of PKCtheta and WASp on symmetry breaking and relocation of the immunological synapse
.
Cell
2007
;
129
:
773
85
.
25.
Brossard
C
,
Feuillet
V
,
Schmitt
A
,
Randriamampita
C
,
Romao
M
,
Raposo
G
, et al
Multifocal structure of the T cell—dendritic cell synapse
.
Eur J Immunol
2005
;
35
:
1741
53
.
26.
Tseng
SY
,
Waite
JC
,
Liu
M
,
Vardhana
S
,
Dustin
ML
. 
T cell-dendritic cell immunological synapses contain TCR-dependent CD28-CD80 clusters that recruit protein kinase C theta
.
J Immunol
2008
;
181
:
4852
63
.
27.
Davis
MM
. 
Molecular genetics of the T cell-receptor beta chain
.
Annu Rev Immunol
1985
;
3
:
537
60
.
28.
Call
ME
,
Pyrdol
J
,
Wiedmann
M
,
Wucherpfennig
KW
. 
The organizing principle in the formation of the T cell receptor-CD3 complex
.
Cell
2002
;
111
:
967
79
.
29.
Reth
M
. 
Antigen receptor tail clue
.
Nature
1989
;
338
:
383
4
.
30.
Irving
BA
,
Weiss
A
. 
The cytoplasmic domain of the T cell receptor zeta chain is sufficient to couple to receptor-associated signal transduction pathways
.
Cell
1991
;
64
:
891
901
.
31.
Blanden
RV
,
Doherty
PC
,
Dunlop
MB
,
Gardner
ID
,
Zinkernagel
RM
,
David
CS
. 
Genes required for cytotoxicity against virus-infected target cells in K and D regions of H-2 complex
.
Nature
1975
;
254
:
269
70
.
32.
Bjorkman
PJ
,
Saper
MA
,
Samraoui
B
,
Bennett
WS
,
Strominger
JL
,
Wiley
DC
. 
Structure of the human class I histocompatibility antigen, HLA-A2
.
Nature
1987
;
329
:
506
12
.
33.
Wakim
LM
,
Bevan
MJ
. 
Cross-dressed dendritic cells drive memory CD8+ T-cell activation after viral infection
.
Nature
2011
;
471
:
629
32
.
34.
Nabavi
N
,
Freeman
GJ
,
Gault
A
,
Godfrey
D
,
Nadler
LM
,
Glimcher
LH
. 
Signalling through the MHC class II cytoplasmic domain is required for antigen presentation and induces B7 expression
.
Nature
1992
;
360
:
266
8
.
35.
Pilones
KA
,
Aryankalayil
J
,
Demaria
S
. 
Invariant NKT cells as novel targets for immunotherapy in solid tumors
.
Clin Dev Immunol
2012
;
2012
:
720803
.
36.
Swain
SL
,
Howard
M
,
Kappler
J
,
Marrack
P
,
Watson
J
,
Booth
R
, et al
Evidence for two distinct classes of murine B cell growth factors with activities in different functional assays
.
J Exp Med
1983
;
158
:
822
35
.
37.
Tadokoro
CE
,
Shakhar
G
,
Shen
S
,
Ding
Y
,
Lino
AC
,
Maraver
A
, et al
Regulatory T cells inhibit stable contacts between CD4+ T cells and dendritic cells in vivo
.
J Exp Med
2006
;
203
:
505
11
.
38.
Tang
Q
,
Adams
JY
,
Tooley
AJ
,
Bi
M
,
Fife
BT
,
Serra
P
, et al
Visualizing regulatory T cell control of autoimmune responses in nonobese diabetic mice
.
Nat Immunol
2006
;
7
:
83
92
.
39.
Leung
MW
,
Shen
S
,
Lafaille
JJ
. 
TCR-dependent differentiation of thymic Foxp3+ cells is limited to small clonal sizes
.
J Exp Med
2009
;
206
:
2121
30
.
40.
Sakaguchi
S
,
Vignali
DA
,
Rudensky
AY
,
Niec
RE
,
Waldmann
H
. 
The plasticity and stability of regulatory T cells
.
Nat Rev Immunol
2013
;
13
:
461
7
.
41.
Zanin-Zhorov
A
,
Lin
J
,
Scher
J
,
Kumari
S
,
Blair
D
,
Hippen
KL
, et al
Scaffold protein disc large homolog 1 is required for T-cell receptor-induced activation of regulatory T-cell function
.
Proc Natl Acad Sci U S A
2012
;
109
:
1625
30
.
42.
Brugnera
E
,
Bhandoola
A
,
Cibotti
R
,
Yu
Q
,
Guinter
TI
,
Yamashita
Y
, et al
Coreceptor reversal in the thymus: signaled CD4+8+ thymocytes initially terminate CD8 transcription even when differentiating into CD8+ T cells
.
Immunity
2006
;
13
:
59
71
.
43.
Garcia
KC
,
Gapin
L
,
Adams
JJ
,
Birnbaum
ME
,
Scott-Browne
JP
,
Kappler
JW
, et al
A closer look at TCR germline recognition
.
Immunity
2012
;
36
:
887
8
.
44.
Van Laethem
F
,
Tikhonova
AN
,
Pobezinsky
LA
,
Tai
X
,
Kimura
MY
,
Le Saout
C
, et al
Lck availability during thymic selection determines the recognition specificity of the T cell repertoire
.
Cell
2013
;
154
:
1326
41
.
45.
Engelhardt
JJ
,
Boldajipour
B
,
Beemiller
P
,
Pandurangi
P
,
Sorensen
C
,
Werb
Z
,. 
Marginating dendritic cells of the tumor microenvironment cross-present tumor antigens and stably engage tumor-specific T cells
.
Cancer Cell
2012
;
21
:
402
17
.
46.
Kantoff
PW
,
Higano
CS
,
Shore
ND
,
Berger
ER
,
Small
EJ
,
Penson
DF
, et al
Sipuleucel-T immunotherapy for castration-resistant prostate cancer
.
N Engl J Med
2010
;
363
:
411
22
.
47.
Porter
DL
,
Levine
BL
,
Kalos
M
,
Bagg
A
,
June
CH
. 
Chimeric antigen receptor-modified T cells in chronic lymphoid leukemia
.
N Engl J Med
2011
;
365
:
725
33
.
48.
Schmits
R
,
Kundig
TM
,
Baker
DM
,
Shumaker
G
,
Simard
JJ
,
Duncan
G
, et al
LFA-1-deficient mice show normal CTL responses to virus but fail to reject immunogenic tumor
.
J Exp Med
1996
;
183
:
1415
26
.
49.
Sanchez-Madrid
F
,
Krensky
AM
,
Ware
CF
,
Robbins
E
,
Strominger
JL
,
Burakoff
SJ
, et al
Three distinct antigens associated with human T-lymphocyte-mediated cytolysis: LFA-1, LFA-2, and LFA-3
.
Proc Natl Acad Sci U S A
1982
;
79
:
7489
93
.
50.
Shaw
S
,
Luce
GE
,
Quinones
R
,
Gress
RE
,
Springer
TA
,
Sanders
ME
. 
Two antigen-independent adhesion pathways used by human cytotoxic T-cell clones
.
Nature
1986
;
323
:
262
4
.
51.
Wild
MK
,
Cambiaggi
A
,
Brown
MH
,
Davies
EA
,
Ohno
H
,
Saito
T
, et al
Dependence of T cell antigen recognition on the dimensions of an accessory receptor-ligand complex
.
J Exp Med
1999
;
190
:
31
41
.
52.
Latour
S
,
Roncagalli
R
,
Chen
R
,
Bakinowski
M
,
Shi
X
,
Schwartzberg
PL
, et al
Binding of SAP SH2 domain to FynT SH3 domain reveals a novel mechanism of receptor signalling in immune regulation
.
Nat Cell Biol
2003
;
5
:
149
54
.
53.
Pasquier
B
,
Yin
L
,
Fondaneche
MC
,
Relouzat
F
,
Bloch-Queyrat
C
,
Lambert
N
, et al
Defective NKT cell development in mice and humans lacking the adapter SAP, the X-linked lymphoproliferative syndrome gene product
.
J Exp Med
2005
;
201
:
695
701
.
54.
Rothlein
R
,
Dustin
ML
,
Marlin
SD
,
Springer
TA
. 
A human intercellular adhesion molecule (ICAM-1) distinct from LFA-1
.
J Immunol
1986
;
137
:
1270
4
.
55.
Staunton
DE
,
Dustin
ML
,
Springer
TA
. 
Functional cloning of ICAM-2, a cell adhesion ligand for LFA-1 homologous to ICAM-1
.
Nature
1989
;
339
:
61
4
.
56.
de Fougerolles
AR
,
Springer
TA
. 
Intercellular adhesion molecule 3, a third adhesion counter-receptor for lymphocyte function-associated molecule 1 on resting lymphocytes
.
J Exp Med
1992
;
175
:
185
90
.
57.
Shimizu
Y
,
van Seventer
GA
,
Horgan
KJ
,
Shaw
S
. 
Roles of adhesion molecules in T-cell recognition: fundamental similarities between four integrins on resting human T cells (LFA-1, VLA-4, VLA-5, VLA-6) in expression, binding, and costimulation
.
Immunol Rev
1990
;
114
:
109
43
.
58.
Dustin
ML
,
Rothlein
R
,
Bhan
AK
,
Dinarello
CA
,
Springer
TA
. 
Induction by IL 1 and interferon-gamma: tissue distribution, biochemistry, and function of a natural adherence molecule (ICAM-1)
.
J Immunol
1986
;
137
:
245
54
.
59.
Dustin
ML
,
Springer
TA
. 
Lymphocyte function-associated antigen-1 (LFA-1) interaction with intercellular adhesion molecule-1 (ICAM-1) is one of at least three mechanisms for lymphocyte adhesion to cultured endothelial cells
.
J Cell Biol
1988
;
107
:
321
31
.
60.
Major
EO
. 
Progressive multifocal leukoencephalopathy in patients on immunomodulatory therapies
.
Annu Rev Med
2010
;
61
:
35
47
.
61.
Park
EJ
,
Peixoto
A
,
Imai
Y
,
Goodarzi
A
,
Cheng
G
,
Carman
CV
, et al
Distinct roles for LFA-1 affinity regulation during T-cell adhesion, diapedesis, and interstitial migration in lymph nodes
.
Blood
2010
;
115
:
1572
81
.
62.
Peterson
EJ
,
Woods
ML
,
Dmowski
SA
,
Derimanov
G
,
Jordan
MS
,
Wu
JN
, et al
Coupling of the TCR to integrin activation by Slap-130/Fyb
.
Science
2001
;
293
:
2263
5
.
63.
Martin
PJ
,
Ledbetter
JA
,
Morishita
Y
,
June
CH
,
Beatty
PG
,
Hansen
JA
. 
A 44 kilodalton cell surface homodimer regulates interleukin 2 production by activated human T lymphocytes
.
J Immunol
1986
;
136
:
3282
7
.
64.
Bromley
SK
,
Iaboni
A
,
Davis
SJ
,
Whitty
A
,
Green
JM
,
Shaw
AS
, et al
The immunological synapse and CD28-CD80 interactions
.
Nat Immunol
2001
;
2
:
1159
66
.
65.
Babcock
SK
,
Gill
RG
,
Bellgrau
D
,
Lafferty
KJ
. 
Studies on the two-signal model for T cell activation in vivo
.
Transplant Proc
1987
;
19
:
303
6
.
66.
McHugh
RS
,
Ahmed
SN
,
Wang
YC
,
Sell
KW
,
Selvaraj
P
. 
Construction, purification, and functional incorporation on tumor cells of glycolipid-anchored human B7-1 (CD80)
.
Proc Natl Acad Sci U S A
1995
;
92
:
8059
63
.
67.
Jenkins
MK
,
Schwartz
RH
. 
Antigen presentation by chemically modified splenocytes induces antigen-specific T cell unresponsiveness in vitro and in vivo
.
J Exp Med
1987
;
165
:
302
19
.
68.
Kong
KF
,
Yokosuka
T
,
Canonigo-Balancio
AJ
,
Isakov
N
,
Saito
T
,
Altman
A
. 
A motif in the V3 domain of the kinase PKC-theta determines its localization in the immunological synapse and functions in T cells via association with CD28
.
Nat Immunol
2011
;
12
:
1105
12
.
69.
Inaba
K
,
Witmer-Pack
M
,
Inaba
M
,
Hathcock
KS
,
Sakuta
H
,
Azuma
M
, et al
The tissue distribution of the B7-2 costimulator in mice: abundant expression on dendritic cells in situ and during maturation in vitro
.
J Exp Med
1994
;
180
:
1849
60
.
70.
Medzhitov
R
,
Preston-Hurlburt
P
,
Janeway
CA
 Jr
. 
A human homologue of the Drosophila Toll protein signals activation of adaptive immunity
.
Nature
1997
;
388
:
394
7
.
71.
Paulos
CM
,
Carpenito
C
,
Plesa
G
,
Suhoski
MM
,
Varela-Rohena
A
,
Golovina
TN
, et al
The inducible costimulator (ICOS) is critical for the development of human T(H)17 cells
.
Sci Transl Med
2010
;
2
:
55ra78
.
72.
Xu
H
,
Li
X
,
Liu
D
,
Li
J
,
Chen
X
,
Hou
S
, et al
Follicular T-helper cell recruitment governed by bystander B cells and ICOS-driven motility
.
Nature
2013
;
496
:
523
7
.
73.
Shambharkar
PB
,
Blonska
M
,
Pappu
BP
,
Li
H
,
You
Y
,
Sakurai
H
, et al
Phosphorylation and ubiquitination of the I kappa B kinase complex by two distinct signaling pathways
.
EMBO J
2007
;
26
:
1794
805
.
74.
So
T
,
Soroosh
P
,
Eun
SY
,
Altman
A
,
Croft
M
. 
Antigen-independent signalosome of CARMA1, PKCtheta, and TNF receptor-associated factor 2 (TRAF2) determines NF-kappaB signaling in T cells
.
Proc Natl Acad Sci U S A
2011
;
108
:
2903
8
.
75.
Giurisato
E
,
Cella
M
,
Takai
T
,
Kurosaki
T
,
Feng
Y
,
Longmore
GD
, et al
Phosphatidylinositol 3-kinase activation is required to form the NKG2D immunological synapse
.
Mol Cell Biol
2007
;
27
:
8583
99
.
76.
Raulet
DH
,
Gasser
S
,
Gowen
BG
,
Deng
W
,
Jung
H
. 
Regulation of ligands for the NKG2D activating receptor
.
Annu Rev Immunol
2013
;
31
:
413
41
.
77.
Kaiser
BK
,
Yim
D
,
Chow
IT
,
Gonzalez
S
,
Dai
Z
,
Mann
HH
, et al
Disulphide-isomerase-enabled shedding of tumour-associated NKG2D ligands
.
Nature
2007
;
447
:
482
6
.
78.
Markiewicz
MA
,
Carayannopoulos
LN
,
Naidenko
OV
,
Matsui
K
,
Burack
WR
,
Wise
EL
, et al
Costimulation through NKG2D enhances murine CD8+ CTL function: similarities and differences between NKG2D and CD28 costimulation
.
J Immunol
2005
;
175
:
2825
33
.
79.
Crites
TJ
,
Padhan
K
,
Muller
J
,
Krogsgaard
M
,
Gudla
PR
,
Lockett
SJ
, et al
TCR Microclusters pre-exist and contain molecules necessary for TCR signal transduction
.
J Immunol
2014
;
193
:
56
67
.
80.
Krummel
MF
,
Allison
JP
. 
CD28 and CTLA-4 have opposing effects on the response of T cells to stimulation
.
J Exp Med
1995
;
182
:
459
65
.
81.
Krummel
MF
,
Allison
JP
. 
CTLA-4 engagement inhibits IL-2 accumulation and cell cycle progression upon activation of resting T cells
.
J Exp Med
1996
;
183
:
2533
40
.
82.
Egen
JG
,
Allison
JP
. 
Cytotoxic T lymphocyte antigen-4 accumulation in the immunological synapse is regulated by TCR signal strength
.
Immunity
2002
;
16
:
23
35
.
83.
Pentcheva-Hoang
T
,
Egen
JG
,
Wojnoonski
K
,
Allison
JP
. 
B7-1 and b7-2 selectively recruit CTLA-4 and CD28 to the immunological synapse
.
Immunity
2004
;
21
:
401
13
.
84.
Qureshi
OS
,
Zheng
Y
,
Nakamura
K
,
Attridge
K
,
Manzolti
C
,
Schmidt
EM
, et al
Trans-endocytosis of CD80 and CD86: a molecular basis for the cell-extrinsic function of CTLA-4
.
Science
2011
;
332
:
600
3
.
85.
Kong
KF
,
Fu
G
,
Zhang
Y
,
Yokosuka
T
,
Casas
J
,
Canonigo-Balancio
AJ
, et al
Protein kinase C-eta controls CTLA-4-mediated regulatory T cell function
.
Nat Immunol
2014
;
15
:
465
72
.
86.
Wolchok
JD
,
Hodi
FS
,
Weber
JS
,
Allison
JP
,
Urba
WJ
,
Robert
C
, et al
Development of ipilimumab: a novel immunotherapeutic approach for the treatment of advanced melanoma
.
Ann N Y Acad Sci
2013
;
1291
:
1
13
.
87.
Kim
JM
,
Rasmussen
JP
,
Rudensky
AY
. 
Regulatory T cells prevent catastrophic autoimmunity throughout the lifespan of mice
.
Nat Immunol
2007
;
8
:
191
7
.
88.
Ishida
Y
,
Agata
Y
,
Shibahara
K
,
Honjo
T
. 
Induced expression of PD-1, a novel member of the immunoglobulin gene superfamily, upon programmed cell death
.
EMBO J
1992
;
11
:
3887
95
.
89.
Latchman
Y
,
Wood
CR
,
Chernova
T
,
Chaudhary
D
,
Borde
M
,
Chernova
I
, et al
PD-L2 is a second ligand for PD-1 and inhibits T cell activation
.
Nat Immunol
2001
;
2
:
261
8
.
90.
Pentcheva-Hoang
T
,
Chen
L
,
Pardoll
DM
,
Allison
JP
. 
Programmed death-1 concentration at the immunological synapse is determined by ligand affinity and availability
.
Proc Natl Acad Sci U S A
2007
;
104
:
17765
70
.
91.
Yokosuka
T
,
Takamatsu
M
,
Kobayashi-Imanishi
W
,
Hashimoto-Tane
A
,
Azuma
M
,
Saito
T
. 
Programmed cell death 1 forms negative costimulatory microclusters that directly inhibit T cell receptor signaling by recruiting phosphatase SHP2
.
J Exp Med
2012
;
209
:
1201
17
.
92.
Riley
JL
. 
PD-1 signaling in primary T cells
.
Immunol Rev
2009
;
229
:
114
25
.
93.
Fife
BT
,
Pauken
KE
,
Eagar
TN
,
Obu
T
,
Wu
J
,
Tang
Q
, et al
Interactions between PD-1 and PD-L1 promote tolerance by blocking the TCR-induced stop signal
.
Nat Immunol
2009
;
10
:
1185
92
.
94.
Honda
T
,
Egen
JG
,
Lammermann
T
,
Kastenmuller
W
,
Torabi-Parizi
P
,
Germain
RN
. 
Tuning of antigen sensitivity by T cell receptor-dependent negative feedback controls T cell effector function in inflamed tissues
.
Immunity
2014
;
40
:
235
47
.
95.
Zinselmeyer
BH
,
Heydari
S
,
Sacristan
C
,
Nayak
D
,
Cammer
M
,
Herz
J
, et al
PD-1 promotes immune exhaustion by inducing antiviral T cell motility paralysis
.
J Exp Med
2013
;
210
:
757
74
.
96.
Barber
DL
,
Wherry
EJ
,
Masopust
D
,
Zhu
B
,
Allison
JP
,
Sharpe
AH
, et al
Restoring function in exhausted CD8 T cells during chronic viral infection
.
Nature
2006
;
439
:
682
7
.
97.
Day
CL
,
Kaufmann
DE
,
Kiepiela
P
,
Brown
JA
,
Moodley
ES
,
Reddy
S
, et al
PD-1 expression on HIV-specific T cells is associated with T-cell exhaustion and disease progression
.
Nature
2006
;
443
:
350
4
.
98.
Topalian
SL
,
Sznol
M
,
McDermott
DF
,
Kluger
HM
,
Carvajal
RD
,
Sharfman
WH
, et al
Survival, durable tumor remission, and long-term safety in patients with advanced melanoma receiving nivolumab
.
J Clin Oncol
2014
;
32
:
1020
30
.
99.
Curran
MA
,
Montalvo
W
,
Yagita
H
,
Allison
JP
. 
PD-1 and CTLA-4 combination blockade expands infiltrating T cells and reduces regulatory T and myeloid cells within B16 melanoma tumors
.
Proc Natl Acad Sci U S A
2010
;
107
:
4275
80
.
100.
Theocharides
AP
,
Jin
L
,
Cheng
PY
,
Prasolava
TK
,
Malko
AV
,
Ho
JM
, et al
Disruption of SIRPalpha signaling in macrophages eliminates human acute myeloid leukemia stem cells in xenografts
.
J Exp Med
2012
;
209
:
1883
99
.
101.
Lines
JL
,
Sempere
LF
,
Broughton
T
,
Wang
L
,
Noelle
R
. 
VISTA is a novel broad-spectrum negative checkpoint regulator for cancer immunotherapy
.
Cancer Immunol Res
2014
;
2
:
510
7
.
102.
Dustin
ML
,
Springer
TA
. 
T cell receptor cross-linking transiently stimulates adhesiveness through LFA-1
.
Nature
1989
;
341
:
619
24
.
103.
Bachmann
MF
,
McKall-Faienza
K
,
Schmits
R
,
Bouchard
D
,
Beach
J
,
Speiser
DE
, et al
Distinct roles for LFA-1 and CD28 during activation of naive T cells: adhesion versus costimulation
.
Immunity
1997
;
7
:
549
57
.
104.
Shimaoka
M
,
Kim
M
,
Cohen
EH
,
Yang
W
,
Astrof
N
,
Peer
D
, et al
AL-57, a ligand-mimetic antibody to integrin LFA-1, reveals chemokine-induced affinity up-regulation in lymphocytes
.
Proc Natl Acad Sci U S A
2006
;
103
:
13991
6
.
105.
Bromley
SK
,
Dustin
ML
. 
Stimulation of naive T-cell adhesion and immunological synapse formation by chemokine-dependent and -independent mechanisms
.
Immunology
2002
;
106
:
289
98
.
106.
Springer
TA
,
Dustin
ML
. 
Integrin inside-out signaling and the immunological synapse
.
Curr Opin Cell Biol
2012
;
24
:
107
15
.
107.
Meuer
SC
,
Hussey
RE
,
Fabbi
M
,
Fox
D
,
Acuto
O
,
Fitzgerald
KA
, et al
An alternative pathway of T-cell activation: a functional role for the 50 kd T11 sheep erythrocyte receptor protein
.
Cell
1984
;
36
:
897
906
.
108.
Evans
EJ
,
Esnouf
RM
,
Manso-Sancho
R
,
Gilbert
RJ
,
James
JR
,
Yu
C
, et al
Crystal structure of a soluble CD28-Fab complex
.
Nat Immunol
2005
;
6
:
271
9
.
109.
Hunig
T
. 
The storm has cleared: lessons from the CD28 superagonist TGN1412 trial
.
Nat Rev Immunol
2012
;
12
:
317
8
.
110.
Bunnell
SC
,
Hong
DI
,
Kardon
JR
,
Yamazaki
T
,
McGlade
CJ
,
Barr
VA
, et al
T cell receptor ligation induces the formation of dynamically regulated signaling assemblies
.
J Cell Biol
2002
;
158
:
1263
75
.
111.
Campi
G
,
Varma
R
,
Dustin
ML
. 
Actin and agonist MHC-peptide complex-dependent T cell receptor microclusters as scaffolds for signaling
.
J Exp Med
2005
;
202
:
1031
6
.
112.
Mossman
KD
,
Campi
G
,
Groves
JT
,
Dustin
ML
. 
Altered TCR signaling from geometrically repatterned immunological synapses
.
Science
2005
;
310
:
1191
3
.
113.
Varma
R
,
Campi
G
,
Yokosuka
T
,
Saito
T
,
Dustin
ML
. 
T cell receptor-proximal signals are sustained in peripheral microclusters and terminated in the central supramolecular activation cluster
.
Immunity
2006
;
25
:
117
27
.
114.
Lillemeier
BF
,
Mortelmaier
MA
,
Forstner
MB
,
Huppa
JB
,
Groves
JT
,
Davis
MM
. 
TCR and Lat are expressed on separate protein islands on T cell membranes and concatenate during activation
.
Nat Immunol
2010
;
11
:
90
6
.
115.
Samelson
LE
,
Patel
MD
,
Weissman
AM
,
Harford
JB
,
Klausner
RD
. 
Antigen activation of murine T cells induces tyrosine phosphorylation of a polypeptide associated with the T cell antigen receptor
.
Cell
1986
;
46
:
1083
90
.
116.
Weiss
A
,
Littman
DR
. 
Signal transduction by lymphocyte antigen receptors
.
Cell
1994
;
76
:
263
74
.
117.
Dustin
ML
,
Chan
AC
. 
Signaling takes shape in the immune system
.
Cell
2000
;
103
:
283
94
.
118.
Guy
CS
,
Vignali
KM
,
Temirov
J
,
Bettini
ML
,
Overacre
AE
,
Smeltzer
M
, et al
Distinct TCR signaling pathways drive proliferation and cytokine production in T cells
.
Nat Immunol
2013
;
14
:
262
70
.
119.
Tan
YX
,
Manz
BN
,
Freedman
TS
,
Zhang
C
,
Shokat
KM
,
Weiss
A
. 
Inhibition of the kinase Csk in thymocytes reveals a requirement for actin remodeling in the initiation of full TCR signaling
.
Nat Immunol
2014
;
15
:
186
94
.
120.
Koretzky
GA
,
Picus
J
,
Thomas
ML
,
Weiss
A
. 
Tyrosine phosphatase CD45 is essential for coupling T-cell antigen receptor to the phosphatidyl inositol pathway
.
Nature
1990
;
346
:
66
8
.
121.
Mustelin
T
,
Williams
S
,
Tailor
P
,
Couture
C
,
Zenner
G
,
Burn
P
, et al
Regulation of the p70zap tyrosine protein kinase in T cells by the CD45 phosphotyrosine phosphatase
.
Eur J Immunol
1995
;
25
:
942
6
.
122.
James
JR
,
Vale
RD
. 
Biophysical mechanism of T-cell receptor triggering in a reconstituted system
.
Nature
2012
;
487
:
64
9
.
123.
Cordoba
SP
,
Choudhuri
K
,
Zhang
H
,
Bridge
M
,
Basat
AB
,
Dustin
ML
, et al
The large ectodomains of CD45 and CD148 regulate their segregation from and inhibition of ligated T-cell receptor
.
Blood
2013
;
121
:
4295
302
.
124.
Irles
C
,
Symons
A
,
Michel
F
,
Bakker
TR
,
van der Merwe
PA
,
Acuto
O
. 
CD45 ectodomain controls interaction with GEMs and Lck activity for optimal TCR signaling
.
Nat Immunol
2003
;
4
:
189
97
.
125.
Yi
J
,
Wu
XS
,
Crites
T
,
Hammer
JA
 3rd
. 
Actin retrograde flow and actomyosin II arc contraction drive receptor cluster dynamics at the immunological synapse in Jurkat T cells
.
Mol Biol Cell
2012
;
23
:
834
52
.
126.
Yokosuka
T
,
Sakata-Sogawa
K
,
Kobayashi
W
,
Hiroshima
M
,
Hashimoto-Tane
A
,
Tokunaga
M
, et al
Newly generated T cell receptor microclusters initiate and sustain T cell activation by recruitment of Zap70 and SLP-76
.
Nat Immunol
2005
;
6
:
1253
62
.
127.
Kaizuka
Y
,
Douglass
AD
,
Varma
R
,
Dustin
ML
,
Vale
RD
. 
Mechanisms for segregating T cell receptor and adhesion molecules during immunological synapse formation in Jurkat T cells
.
Proc Natl Acad Sci U S A
2007
;
104
:
20296
301
.
128.
Hartman
NC
,
Nye
JA
,
Groves
JT
. 
Cluster size regulates protein sorting in the immunological synapse
.
Proc Natl Acad Sci U S A
2009
;
106
:
12729
34
.
129.
Choudhuri
K
,
Llodra
J
,
Roth
EW
,
Tsai
J
,
Gordo
S
,
Wucherpfennig
KW
, et al
Polarized release of T-cell-receptor-enriched microvesicles at the immunological synapse
.
Nature
2014
;
507
:
118
23
.
130.
Dustin
ML
. 
What counts in the immunological synapse?
Mol Cell
2014
;
54
:
255
62
.
131.
Mittelbrunn
M
,
Gutierrez-Vazquez
C
,
Villarroya-Beltri
C
,
Gonzalez
S
,
Sanchez-Cabo
F
,
Gonzalez
MA
, et al
Unidirectional transfer of microRNA-loaded exosomes from T cells to antigen-presenting cells
.
Nat Commun
2011
;
2
:
282
.
132.
Sage
PT
,
Varghese
LM
,
Martinelli
R
,
Sciuto
TE
,
Kamei
M
,
Dvorak
AM
, et al
Antigen recognition is facilitated by invadosome-like protrusions formed by memory/effector T cells
.
J Immunol
2012
;
188
:
3686
99
.
133.
Suzuki
J
,
Yamasaki
S
,
Wu
J
,
Koretzky
GA
,
Saito
T
. 
The actin cloud induced by LFA-1-mediated outside-in signals lowers the threshold for T-cell activation
.
Blood
2007
;
109
:
168
75
.
134.
Singleton
KL
,
Gosh
M
,
Dandekar
RD
,
Au-Yeung
BB
,
Ksionda
O
,
Tybulewicz
VL
, et al
Itk controls the spatiotemporal organization of T cell activation
.
Sci Signal
2011
;
4
:
ra66
.
135.
Purbhoo
MA
,
Liu
H
,
Oddos
S
,
Owen
DM
,
Neil
MA
,
Pageon
SV
, et al
Dynamics of subsynaptic vesicles and surface microclusters at the immunological synapse
.
Sci Signal
2010
;
3
:
ra36
.
136.
Williamson
DJ
,
Owen
DM
,
Rossy
J
,
Magenau
A
,
Wehrmann
M
,
Gooding
JJ
, et al
Pre-existing clusters of the adaptor Lat do not participate in early T cell signaling events
.
Nat Immunol
2011
;
12
:
655
62
.
137.
Geiger
B
,
Rosen
D
,
Berke
G
. 
Spatial relationships of microtubule-organizing centers and the contact area of cytotoxic T lymphocytes and target cells
.
J Cell Biol
1982
;
95
:
137
43
.
138.
Kupfer
A
,
Swain
SL
,
Janeway
CA
 Jr
,
Singer
SJ
. 
The specific direct interaction of helper T cells and antigen-presenting B cells
.
Proc Natl Acad Sci U S A
1986
;
83
:
6080
3
.
139.
Poo
WJ
,
Conrad
L
,
Janeway
CA
 Jr
. 
Receptor-directed focusing of lymphokine release by helper T cells
.
Nature
1988
;
332
:
378
80
.
140.
Huse
M
,
Lillemeier
BF
,
Kuhns
MS
,
Chen
DS
,
Davis
MM
. 
T cells use two directionally distinct pathways for cytokine secretion
.
Nat Immunol
2006
;
7
:
247
55
.
141.
Das
V
,
Nal
B
,
Dujeancourt
A
,
Thoulouze
MI
,
Galli
T
,
Roux
P
, et al
Activation-induced polarized recycling targets T cell antigen receptors to the immunological synapse; involvement of SNARE complexes
.
Immunity
2004
;
20
:
577
88
.
142.
Finetti
F
,
Paccani
SR
,
Riparbelli
MG
,
Giacomello
E
,
Perinetti
G
,
Pazour
GJ
, et al
Intraflagellar transport is required for polarized recycling of the TCR/CD3 complex to the immune synapse
.
Nat Cell Biol
2009
;
11
:
1332
9
.
143.
de la Roche
M
,
Ritter
AT
,
Angus
KL
,
Dinsmore
C
,
Earnshaw
CH
,
Reiter
JF
, et al
Hedgehog signaling controls T cell killing at the immunological synapse
.
Science
2013
;
342
:
1247
50
.
144.
Bertrand
F
,
Muller
S
,
Roh
KH
,
Laurent
C
,
Dupre
L
,
Valitutti
S
. 
An initial and rapid step of lytic granule secretion precedes microtubule organizing center polarization at the cytotoxic T lymphocyte/target cell synapse
.
Proc Natl Acad Sci U S A
2013
;
110
:
6073
8
.
145.
Deguine
J
,
Breart
B
,
Lemaitre
F
,
Di Santo
JP
,
Bousso
P
. 
Intravital imaging reveals distinct dynamics for natural killer and CD8(+) T cells during tumor regression
.
Immunity
2010
;
33
:
632
44
.
146.
Deguine
J
,
Breart
B
,
Lemaitre
F
,
Bousso
P
. 
Cutting edge: tumor-targeting antibodies enhance NKG2D-mediated NK cell cytotoxicity by stabilizing NK cell-tumor cell interactions
.
J Immunol
2012
;
189
:
5493
7
.
147.
Beal
AM
,
Anikeeva
N
,
Varma
R
,
Cameron
TO
,
Norris
PJ
,
Dustin
ML
, et al
Protein kinase C{theta} regulates stability of the peripheral adhesion ring junction and contributes to the sensitivity of target cell lysis by CTL
.
J Immunol
2008
;
181
:
4815
24
.
148.
Beal
AM
,
Anikeeva
N
,
Varma
R
,
Cameron
TO
,
Vasiliver-Shamis
G
,
Norris
PJ
, et al
Kinetics of early T cell receptor signaling regulate the pathway of lytic granule delivery to the secretory domain
.
Immunity
2009
;
31
:
632
42
.
149.
Ruocco
MG
,
Pilones
KA
,
Kawashima
N
,
Cammer
M
,
Huang
J
,
Babb
JS
, et al
Suppressing T cell motility induced by anti-CTLA-4 monotherapy improves antitumor effects
.
J Clin Invest
2012
;
122
:
3718
30
.
150.
Schneider
H
,
Downey
J
,
Smith
A
,
Zinselmeyer
BH
,
Rush
C
,
Brewer
JM
, et al
Reversal of the TCR stop signal by CTLA-4
.
Science
2006
;
313
:
1972
5
.
151.
Martinez-Martin
N
,
Fernandez-Arenas
E
,
Cemerski
S
,
Delgado
P
,
Turner
M
,
Heuser
J
, et al
T cell receptor internalization from the immunological synapse is mediated by TC21 and RhoG GTPase-dependent phagocytosis
.
Immunity
2011
;
35
:
208
22
.
152.
Huang
JF
,
Yang
Y
,
Sepulveda
H
,
Shi
W
,
Hwang
I
,
Peterson
PA
, et al
TCR-mediated internalization of peptide-MHC complexes acquired by T cells
.
Science
1999
;
286
:
952
4
.
153.
Peters
PJ
,
Geuze
HJ
,
Van der Donk
HA
,
Slot
JW
,
Griffith
JM
,
Stam
NJ
, et al
Molecules relevant for T cell-target cell interaction are present in cytolytic granules of human T lymphocytes
.
Eur J Immunol
1989
;
19
:
1469
75
.
154.
Peters
PJ
,
Geuze
HJ
,
van der Donk
HA
,
Borst
J
. 
A new model for lethal hit delivery by cytotoxic T lymphocytes
.
Immunol Today
1990
;
11
:
28
32
.