Abstract
Advances in academic and clinical studies during the last several years have resulted in practical outcomes in adoptive immune therapy of cancer. Immune cells can be programmed with molecular modules that increase their therapeutic potency and specificity. It has become obvious that successful immunotherapy must take into account the full complexity of the immune system and, when possible, include the use of multifactor cell reprogramming that allows fast adjustment during the treatment. Today, practically all immune cells can be stably or transiently reprogrammed against cancer. Here, we review works related to T cell reprogramming, as the most developed field in immunotherapy. We discuss factors that determine the specific roles of αβ and γδ T cells in the immune system and the structure and function of T cell receptors in relation to other structures involved in T cell target recognition and immune response. We also discuss the aspects of T cell engineering, specifically the construction of synthetic T cell receptors (synTCRs) and chimeric antigen receptors (CARs) and the use of engineered T cells in integrative multifactor therapy of cancer.
Keywords: T cell, T cell receptor (TCR), Chimeric antigen receptor (CAR), alpha beta T cells, gamma delta T cells, Memory T cells, Immune synapse, Reprogramming, Adoptive cell therapy, Signal transduction, TCR clustering
1. Introduction
Progress in immunotherapy has reached a critical point where available funding and efforts can provide practical improved clinical outcomes for patients. These advances are based on findings in academic and clinical studies in immunology, adoptive immunotherapy, gene editing, and stem cell modulation, among other fields. Despite our rapidly increasing understanding of tumor–immune system interactions, there are profound limits to our knowledge. Nonetheless, the urgent need for therapeutic improvements facilitates the development of new drugs and modified cells in parallel with new methods of their clinical evaluation. Particularly important is the opportunity to exploit combinatorial multifactor treatment protocols based on protein and cell engineering.
Immune cells can be programmed with molecular modules that increase their therapeutic potency and specificity. Although in its infancy, modern immunotherapy strives to provide personalized therapy that is modifiable during the course of treatment based on the patient’s baseline characteristics and ongoing accurate evaluation of the course of the disease. The development of immune modulation by cell reprogramming already has been translated into patient cures. This highlights both a fascinating discovery and our relative ignorance about how to prevent high morbidity, off-target effects, and other complications.
Fortunately, some of the gaps in our knowledge are being addressed rapidly. It has become obvious that successful immunotherapy must take into account the full complexity of the immune system and when possible include the use of different types of immunocytes and multifactor cell reprogramming, and apply flexible methods that allow fast adjustment during the treatment depending on the patient’s conditions and needs. Today, practically all immune cells can be stably or transiently reprogrammed against cancer. The most developed field is T cell reprogramming, although quite promising results have been achieved with natural killer (NK) cells, macrophages, and others [1–5]. Several chapters in this volume exemplify these different sources of cells that can be reprogrammed (e.g., Chapters 6–9 for NK cells, Chapter 11 for dendritic cells (DCs), Chapter 14 for macrophages).
Challenges in cell engineering appear at many levels. At the subcellular level, the design of novel proteins or other molecules that can be expressed and function in accord with endogenous cell systems is nontrivial. At the cellular level, the complexity of cell-to-cell interaction dictates accurate construct adjustment and modification. In addition, there may be the need to introduce additional molecules of different classes that optimize the cognate cell function. At the organismal level, there is a need to evaluate multiple reactions by an integral combinatorial approach where cell engineering synergistically couples with other therapies.
During last 20 years, adoptive cell therapy (ACT) was developed based on two main premises: (1) Cytotoxic T cells eliminate diseased cells, and (2) artificial modular protein constructs can be designed to recognize specific antigens on the surface of target cells and trigger T cell target killing. Since 2011, the number of patents related to chimeric antigen receptor (CAR)-mediated immunotherapy has grown exponentially [6]. Today, ACT is best demonstrated in the treatment of blood B cell tumors with chimeric antigen receptor T cell (CAR-T) therapy products: Kymriah (Novartis) and Yescarta (Kite Pharma/Gilead), which are approved by the US Food and Drug Administration [7] and the European Medicines Agency [8]. Other hematopoietic cancers and solid cancers have been more challenging to target, because T cell function is impeded by the absence of specific tumor antigens, multiple barriers of tumor accessibility, and immunosuppressive conditions. Increased knowledge of the processes that take place in the tumor microenvironment, metastasis development, and immune tuning on both systemic and local levels will be necessary to improve cell engineering and resolve both the fundamental and technical problems. In this review, we briefly address the main areas in T cell reprogramming relevant to ACT of cancer and describe some obvious underdeveloped areas important for building better integrative personalized therapies.
2. T Cells
2.1. T Cell Diversity
Functional diversity of T cell populations and their T cell receptor (TCR) repertoire are important factors that determine immune health. In cancer research, attention is often focused on the rather narrow task of finding a T cell population, among a weakened immune system, that is robust enough to yield a sufficient amount and be reprogrammed to kill cancer targets and then maintained in patients long enough to achieve efficacy. However, cross-communication among the subsets of T cells, dendritic cells (DCs), and other immunocytes is also an important part of immune response. That explains growing attention to various subsets of immune cells.
In humans, it is estimated that ~7 billion T cells are present in the peripheral blood, 25 billion in the bone marrow, 30 billion in the spleen, and 150 billion in the lymph nodes. Taken together, these three organs contain >200 × 109 T cells, which constitutes the majority of total T cells [9].
T cells consist of many subtypes, the largest of which are the “conventional” αβ T cells with “classic” major histocompatibility complex (MHC) restriction. These T cells are part of the sophisticated adoptive immune system with a relatively slow response. Some other T cells are part of the innate immune system. They are characterized by a limited TCR diversity, are either “non-classic MHC” restricted or MHC independent, and exhibit a fast immune response. They include γδ T cells, natural killer T (NKT) cells, CD1- and MHC class Ib-restricted T cells, MR-1-restricted mucosal-associated invariant T cells (MAIT), and intraepithelial lymphocytes (IELs) [10–12]. Although the subtypes of T cells are functionally different, this difference is not inflexible. For example, human peripheral γδ T cells can be transdifferentiated ex vivo into αβ T cells [13]. γδ T cells are a minor subset of peripheral lymphocytes in humans (<5%) [14] but are enriched in epithelial and mucosal tissues [15].
2.2. T Cell Development
T cells originate in the bone marrow, and most of them develop in the thymus. These cells arise from immature CD4 and CD8 double-negative thymocytes and express either the αβ or γδ TCR [14, 16]. αβ T cells undergo positive and negative selection through recognition of self-peptide–MHC (p–MHC). Most αβ T cells with high affinity to self-peptides undergo negative selection and die. However, some of them survive and after positive selection form certain subtypes, such as IELs, NKTs, and Foxp3+ regulatory T cells (Tregs) [17]. αβ T cells with low affinity to self-p–MHC develop into “conventional” CD8+ cytotoxic lymphocytes (CTL) or CD4+ helper T cells that recognize foreign or “diseased” peptide antigens presented by MHC class I and class II molecules, respectively. This difference is not rigid, as a substantial part of CD4+ T cells can be cytotoxic similar to CD8+ T cells [18–22].
After selection in the thymus, αβ T cells proceed to the periphery where they circulate as naïve CD4+ and CD8+ T cells between blood and secondary lymphoid organs. Positive selection of naïve T cells continues as cyclical “tonic” activation as they circulate between the blood and secondary lymphoid organs where they encounter self-p–MHC [23–25].
γδ T cells recognize antigens in a “classic MHC”-independent manner. They recognize a range of structurally different moieties, such as non-classic MHC molecules, proteins, peptides, and phospholipids [26]. Studies in mice have demonstrated that γδ T cells undergo negative selection in thymus, but the role of positive selection in their development may not be obligatory [27].
2.3. Naïve, Effector, and Memory αβ T Cells
Naïve T cells are maintained in a state of quiescence, which is characterized by low cell volume, low metabolic rate, and low homeostatic proliferation. Gradual naïve T cell propagation without differentiation is upheld by IL7 signaling and tonic, low-level activity of the TCR that monitors “normal” self-p–MHC presented by surrounding cells [28, 29]. Conventional activation of naïve T cells by antigen-presenting cells (APCs) requires the simultaneous engagement of three receptors on the T cells: TCR ligation with agonist p–MHC, CD28 or another costimulatory receptor ligation with cognate ligand, and cytokine receptor ligation with IL2 or another homeostatic cytokine [30–32].
Sometimes, naïve T cells can be activated without TCR ligation. For example, in vivo T cells become activated when their population is severely depleted [33], whereas in vitro T cells can be activated in the presence of high concentrations of interleukins IL2 or IL7 [34].
Upon APC-mediated activation, naïve T cells undergo clonal expansion and differentiation. They activate mTORC1 [35], shift from catabolic to anabolic metabolism, increase glucose and amino acid (AA) uptake, remodel their mitochondrial function, and increase their cell volume and their rate of proliferation [29]. In the initial expansion phase, naïve T cell doubling time decreases from 500–1000 days [36, 37] to a few hours [38, 39]. This process may occur in different tissues and is highly dependent on receptor–ligand affinity and density, as well as environmental factors, such as pH, redox potential, and the availability of nutrients. The activation leads to clonal expansion by sequential asymmetric cell division. In this process, cell divisions convert activated naïve T cells into terminally differentiated effector T cells (TEFF) and self-renewing memory lineages (TMEM) [40]. Some of the progeny cells become more activated and directed toward TEFF, while the others become less metabolically active TMEM [41]. Thus, TMEM cells are metabolically more active than naïve T cells, with higher mitochondrial mass and respiratory capacity [42], but less active than TEFF cells and maintain predominately catabolic metabolism [43]. This contributes to the prolonged persistence of TMEM cells, such that their longevity is between naïve and effector T cells.
Activated TEFF cells include both CD8+ CTLs and CD4+ helper T cells that provide acute protection from immune challenges [44]. Activated CTLs recognize an appropriate target, bind to it, and secrete killing molecules toward the target, before detaching from the dying target [45]. In humans, the main mechanism of T cell cytotoxicity is the granzyme–perforin pathway [46–49]. Other killing systems, such as Fas/FasL, TNF-α/TNF receptor 1, and TRAIL/DR4/DR5, are usually employed in T cell activation-induced cell death (AICD), which is an essential part of T cell homeostasis regulation [50]. However, these systems can also cause target killing [45, 51, 52].
After the immediate immune response and foreign antigen is cleared, the T cell population enters a contraction phase, and most of the TEFF subsequently die by apoptosis [53].
Long-term immune protection is provided by subsets of TMEM cells that include central memory (TCM) and effector memory (TEM) cells. TCM are circulating cells that are prevalent in lymph nodes and have enhanced longevity and proliferative potential. Although TCM lack effector functions themselves, they generate TEFF and TEM cells. TEM cells are circulating cells that are more prevalent in nonlymphoid tissues [54–57]. They possess immediate effector functions; they rapidly migrate toward targets and provide antigen elimination [58]. Differentiation from naïve T cells, to TCM, to TEM is associated with decreasing expression of the Wnt/β catenin transcription factors, LEF-1, and TCF-1 [59–61]. Clonal development is a flexible process, and to some degree, it can be reversed. For example, in mice, long-lived memory CD8+ TCM cells can develop from effector TEFF cells through a process of dedifferentiation [62].
TMEM population is diversified, and their discovery and classification are especially difficult if they are not circulating. Recently, such tissue-resident memory (TRM) cells were located in the lungs, salivary glands, female reproductive tract, skin, and liver, where they orchestrate the response to different pathogens [63, 64].
An important subset within the TCM population generated during the primary immune response has stem cell-like characteristics, defined as TSCM. These cells exhibit a high capacity for homeostatic proliferation and can give rise to other memory subsets. In normal homeostasis, the pool of TSCM cells in humans is believed to comprise 2–3% of all circulating T lymphocytes [61, 65].
Some CD8+ TMEM cells are virtual memory cells that originate from naïve T cells under strong IL15 signaling but without agonist antigen stimulation [66–68]. The role of these cells is debatable, but one theory is that they provide innate-like protection during the earliest stages of bacterial or viral infection [68, 69].
Once created, conventional TMEM cells also become independent of further antigenic stimulation, which allows their maintenance after resolving an acute infection. TMEM homeostasis depends on paracrine and autocrine IL15 signaling, and TMEM cells can proliferate in response to IL15 in a TCR-independent fashion [70, 71]. Because the overall number of TMEM cells remains constant over long periods of time, the observed continual proliferation of TMEM cells must be accompanied by a nearly equal death rate, probably as result of asymmetric division, where half of the offspring becomes apoptotic [72].
2.4. Persistence of Circulating αβ T Cells
The accurate evaluation of T cell subpopulations is difficult and varies in different studies. Vrisekoop et al. estimated that human naïve CD4 and CD8 T cells have half-lives of 4.2 and 6.5 years, respectively, whereas memory CD4 and CD8 T cells have half-lives of 0.4 and 0.7 years [73]. However, these measurements were made on circulating cells, whereas TMEM in nonlymphoid tissues and bone marrow can have much longer life spans that allow long clonal survival. In fact, some memory T cells persist for more than 10 years [74, 75].
The age-related decline in T cell population is caused by thymic involution, impaired peripheral T cell maintenance, repeated antigen exposure, and persistent inflammation. Although healthy aging individuals can maintain a sufficient T cell content over time, naïve CD4+ and CD8+ T cell numbers and repertoire gradually decline [76], and the peripheral T cell pool becomes dominated by memory T cells [77, 78].
3. The T Cell Receptor (TCR)
3.1. Formation of the αβ TCR
Present on more than 90% of the T cells, the αβ TCR comprises the predominant TCR complex on the surface of human T cells [16]. Interestingly, it is also present on some neutrophils [79, 80] and macrophages [81, 82].
To date, no intrinsic enzymatic activity has been found for any of the TCR proteins, but rather, it is provided by the set of cytosolic, transmembrane (TM), and membrane-bound (e.g., myristoylated) enzymes that associate with the TCR. These proteins are organized in a highly cooperative system where the inter-protein signaling is quite often regulated by phosphorylation–dephosphorylation. Other interactions include a variety of enzymatic reactions and cytoskeleton and membrane rearrangements, which result in the attraction of scaffold proteins and adaptors, and signal amplification through multiple downstream metabolic pathways.
The conventional αβ TCR is located in membrane lipid raft subdomains. It contains two antigen recognition proteins, α chain and β chain, and four “signal transduction” CD3 proteins, δ, γ, ε, and ζ. The α and β subunits, bound together by a disulfide bridge, are sequence-variable proteins, with very short cytoplasmic tails, that can recognize agonist peptide in context of the MHC complex. The αβ heterodimer is flanked by the CD3 proteins as non-covalently associated heterodimers of εγ, εδ, and a disulfide-linked homodimer of ζ- [83–85]. The CD3 proteins are invariant, with relatively longer cytosolic domains that contain binding sites for cytosolic adaptors and enzymes, and maintain an invariant path for information of TCR/p–MHC recognition through the cell membrane down to the cytoplasm, to achieve an adequate cellular response [86].
Subunit cooperativity in the TCR is substantially determined by coordination of ionizable residues in their transmembrane (TM) domains, which form a specific hydrophobic/ionic interface between αβ, δε, γε, and ζζ [87]. Each of the ζ, ε, and δ molecules possesses an ionizable aspartic acid in their TM regions, while γ possesses a glutamic acid residue. Together, this gives six acidic residues in the TM region of the TCR complex. The α TM domain possesses two basic residues (arginine and lysine), while the β TM domain possesses one basic residue (lysine) [88]. Therefore, an αβγεδεζζ complex has three extra negative charges in the TM region leading to a “charge imbalance.” This led to a search for an alternative bivalent TCR structure, γεαβζζαβδε, with a neutral TM region [89, 90] or even structures with higher valency [91–93]. Purification of the TCR with different detergents or without detergents has led to various deduced TCR structures [16, 90, 92]. In fact, it is possible that the actual TCR structure may “resonate” between mono-, di-, and higher variants of valency.
TCR proteins emerge on the plasma membrane as a fully built complex. ζ is the last subunit to be associated with TCR complex in the Golgi [16, 88]. In resting human T cells, a portion of ζ associates with the actin cytoskeleton. This interaction, mediated by a sequence in the C-terminus of ζ, might be involved in the localization of the TCR into lipid raft structures as well as play a role in TCR recycling [94]. In the cytosol, ζ is present in excess, compared to the other TCR proteins, and can participate in reactions not related to TCR activity. For example, ζ interacts with the transferrin receptor (TfR), and a TfR-ζ complex is expressed on the cell surface independently from TCR. ζ is also expressed in cells other than T cells, like NK cells and neurons. In NK cells, ζ is associated with NK FcgRIII (CD16) and may participate in cell surface expression of this receptor complex [95]. ζ is also associated with NKp46 and NKp30 receptors on NK cells, and its phosphorylation is required for transmission of activating signals upon antigen binding to these receptors. ζ expression in retinal ganglion cells and brain neurons regulates neuronal development by reducing the size of the dendritic arbor [96].
3.2. TCR/p–MHC Ligation
Upon TCR/p–MHC ligation, the shift in αβ conformation determines transmission of antigen-binding energy on the cell surface down into the CD3 intracellular tails [97]. A plausible interpretation of the available data is that the signaling domains of CD3 in the inactive TCR are submersed in the inner leaflet of the cell membrane which screens them from cytosolic adaptors [16, 83, 98, 99]. The mechanical force applied during TCR/p–MHC ligation weakens intra-subunit associations, changes membrane composition, and leads to subunit rearrangement, moving the CD3 intracellular domains out of the membrane and exposing them to cytosolic signaling adaptors [83, 98, 100–102].
CD3 proteins bind cytosolic adaptors by specific docking motifs. One such motif, tyrosine-based activation motifs (ITAM), is present as a single copy in γ, δ, and ε and as three copies in ζ, for a total of ten ITAMs in the CD3 complex [16]. This distinguishes the TCR from other cell receptors with ITAMs, which contain only one or two. A plausible view on ITAMs’ composition is that ITAM tyrosines in CD3 are not redundant and might have specific roles [103]. However, the exact role of each ITAM in the CD3 subunits, including the detailed dynamics of ITAM phosphorylation, remains to be elucidated.
ITAM multiplicity determines the correct signaling for T cells developing in the thymus [104]. Decreasing the number of CD3 ITAMs to less than seven in mice impedes T cell development. These animals developed autoimmune disease probably due to inefficient signaling under negative selection in the thymus [104]. In these animals, studied prior to the onset of autoimmunity, CD3s with slightly decreased numbers of ITAMs still activated signaling cascades including cytokine production and T cell proliferation. Further decrease of CD3 ITAM multiplicity to two to four ITAMs per CD3 resulted in only a limited response with cytokine production, but not T cell proliferation [105].
For developed T cells, ITAM multiplicity appears to have minimal influence on signal amplitude [106]. Instead, it might help with the coordinated activation of all the subunits in the CD3 complex, such that there is a switch-like “all or nothing” response upon TCR binding to p–MHC [107]. Therefore, ITAM multiplicity may determine potency for synchronous activation of the T cell population [106, 107].
The ITAM motif (YXXL/IX6–8YXXL/I) contains two tyrosines, which are usually phosphorylated by the key TCR activator, lymphocyte-specific protein tyrosine kinase (Lck). Lck associates with CD3 proteins as well as with the coreceptors CD4 and CD8 [108]. Lck exists in open (active) and closed (inactive) forms, which is determined by the state of phosphorylation. The transmembrane phosphatase CD45 activates or inhibits Lck, depending on location of dephosphorylation. Lck can also be inhibited by the cytosolic kinase Csk or by the cytosolic phosphatase SHP-1. Also, Lck can be activated through serine/threonine phosphorylation by extracellular signal-regulated kinase (ERK), and this may interfere with SHP-1 recruitment to the TCR complex [109].
Phosphorylation of both tyrosines in an ITAM by Lck attracts the cytosolic adaptor ZAP-70. After TCR/p–MHC ligation, ζ phosphorylation proceeds from the most membrane-distal ITAM toward the membrane [110], and their affinity to Zap-70 increases in the same direction [111]. When bound to the CD3 ITAM, Zap-70 is not activated, but released from its autoinhibited conformation [112]. This opens Zap-70 to be activated through phosphorylation by Lck or by autophosphorylation [106]. Zap-70 phosphorylation can occur by Lck or Zap-70 molecules that are bound to the same protein or by those associated with neighboring CD3 subunits [106], thereby enhancing CD3 cooperativity.
Two CD3 proteins, ε and ζ, deserve special attention. Both are non-glycosylated proteins and present in the TCR as two copies: ε as heterodimers with δ and γ and ζ as a homodimer with the possibility to undergo covalent S–S binding. ε and ζ subunits contain other docking sites besides ITAMs for cytosolic adaptors and can be involved in regulation beyond the ITAM–Zap-70 reaction [113–115]. For example, ε contains a proline-rich motif that can bind the adaptor protein Nck [116]. Nck participates in actin reorganization, cell adhesion, and movement. Nck binds to a partially phosphorylated ε ITAM, which contains a non-phosphorylated Y39 and a phosphorylated Y50. The Nck–ε interaction peaks in the beginning of TCR/p–MHC ligation and then decreases after 10 min as the phosphorylation of ε increases [103]. The ζ subunit has the longest cytosolic tail protein among the CD3 proteins and the highest involvement in multiplex signaling. It can bind adaptor proteins Shc and Grb2, the p85 subunit of PI3K [86], SLP-76, Vav, and negative regulators such as SLAP [117], SLAP-2 [118, 119], TRIM, CTLA4, and Unc119, as well as actin [113]. These proteins as well as GADS, phospholipase PLCγ1, Nck, p38 MAPK, and ADAP are recruited into a complex with linker for the activation of T cells (LAT) upon its phosphorylation by ZAP-70 [16, 120, 121].
The ensuing reactions may lead to signal amplification or abruption, depending on multiple factors involved in the regulation of T cell activity. When the TCR senses self-p–MHC, it enters a state of “tonic activity” with only partial phosphorylation of CD3 ITAMs and the associated cytosolic adaptors. The weak reaction on p–MHC may trigger a negative feedback, through recruitment of Csk, CD45, and SHP-1, which inactivate Lck and lead to receptor desensitization. In contrast, a stronger reaction may turn on a positive feedback, involving further activation and phosphorylation by Lck and prevention of SHP-1 and CD45 recruitment [109]. The activation triggers further signaling in local and distant protein networks and recruitment of the TCR coreceptors, resulting in a full cellular response [122–124].
3.3. TCR Coreceptors
Upon binding agonist p–MHC on target cell, the αβ TCR attracts transmembrane TCR coreceptors CD8 or CD4 to the p–MHC and checkpoint receptors that recognize additional non-MHC proteins on the target cell membrane. These additional interactions can either augment or undermine TCR signaling outcome.
Coreceptors CD4 or CD8 bind to the p–MHC cooperatively with the TCR, which may increase the overall binding force. However, the most important role of CD4 and CD8 proteins is to deliver activated Lck, associated with coreceptors, into the area of the TCR/p–MHC interaction so that CD3 cytoplasmic tails move out of membrane, become exposed to cytosol, and can be phosphorylated, to attract Zap-70 [100, 125]. Whereas the CD4 receptor is a single protein that spans the cell membrane and binds Lck [126], the CD8 receptor contains two proteins: CD8α and CD8β. While both CD8α and CD8β span the cell membrane, only CD8α binds Lck. Thus, it is likely that CD8α is involved in both ligand recognition and signaling, and CD8β participates only in the recognition. The CD8αβ heterodimer is expressed on CD8 T cells [127]; the CD8αα homodimer is expressed on some γδ T cells, NK cells, and IELs; and the CD8ββ homodimer has not been found on lymphocytes [100]. The binding affinity of CD8 to MHC is independent of TCR specificity or affinity; therefore, the impact of coreceptors on p–MHC binding and signaling decreases with increasing TCR affinity [128].
Coreceptor involvement in TCR/p-MHC ligation is followed by Zap-70-mediated phosphorylation of the scaffold protein LAT, which in turn nucleates multiple downstream pathways. Phosphorylated LAT binds additional adaptors GADS, GRB2, and phospholipase PLCγ1(NF-kB). This LAT complex subsequently recruits other adapters and enzymes, including SLP76, VAV1, Nck, p38 MAPK, and ADAP [16, 120, 121]. Phosphorylated ZAP-70 also has a noncatalytic function as a scaffold phosphoprotein that facilitates the high-affinity state of the integrin LFA-1, which in turn increases T cell adhesion by binding ICAM-1 on antigen-presenting cells [129].
Further T cell activation turns on a cascade of reactions, including structural changes with actin rearrangement [130], attraction of the centrosome, increasing of endocytosis, and further accumulation of TCR receptors in the synapse by lateral diffusion or exocytosis (see also Subheading 3.6). In addition, there is inositol phospholipid hydrolysis and mobilization of Ca2+ through activation of phospholipase C-gamma 1 and serine/threonine kinases [120]. Finally, distant signaling pathways are induced including PI3K/Akt/mTOR, Myc [44, 105, 131–133], NFAT [134], NF-κB, and AP-1 [135]. Overall, the signal cooperativity of CD3 proteins with the coreceptors may include cross-phosphorylation among ITAMs, synergism in adaptors’ binding, and cross-activation among CD3 complexes in TCR clusters.
3.4. The Immune Synapse
The structure and specific activity of immune synapses are determined by the type of T cells (cytotoxic, helper, Treg, NKT), TCR (αβ TCR and γδ TCR), coreceptors (CD4 or CD8), and the set of checkpoint receptors that bind to various ligands outside the p–MHC and add either positive or negative cooperativity. For example, the synapse between a helper CD4+ T cell and B cell exists longer and leads to different outcomes than the synapse between a cytotoxic CD8+ T cell and B cell [136]. As a second example, the synapse with DCs primes naïve CD8+ T cells to proliferate and differentiate into CTLs over the course of several days, whereas it primes CTLs to kill diseased cells by secretion of cytolytic granules at the point of TCR signaling [137].
Target cells also determine synapse structure and function. Potential target cells include “professional” APCs, such as a dendritic cell (DC), macrophage, or B cell [138]; “atypical” APCs, such as a granulocyte [139], lymphatic epithelial cell [140, 141], basophil, mast cell, or eosinophil [138]; or “true target” diseased cells that should be eliminated. Synapses between T cells and different APCs have different organizations [142, 143]. CTLs attached to dendritic cells are less toxic toward their target than CTLs attached to B cells [144, 145].
Wild-type TCRs usually have low affinity for their p–MHC targets with a dissociation equilibrium constant (KD) of 1–100 μM [136, 146, 147]. The precise number of p–MHC target antigens per cell required for optimal αβ T cell activation can vary, but in principle, T cells can be activated in response to only a very few p–MHC antigens [148–151]. In fact, the αβ TCR accurately recognizes relatively infrequent agonist peptides among “self”-peptides in the MHC class I, with a ratio as low as ~1 × 104 self-peptides: one agonist peptide [152]. Technically, it occurs by constant p–MHC monitoring that provides low-level T cell tonic stimulation. It has been proposed that naïve T cells have the ability to adjust their activation threshold by a mechanism dependent not only on specific TCR affinity to p–MHC [153], but includes dynamic tuning with participation of coreceptors CD8 and CD4 and proteins with TCR inhibitory activity, like CD5, CD6, and CD45 [153, 154]. Intrinsic TCR affinity for self-p–MHC ligands in mice correlates with expression of CD5 [154, 155] and inversely correlates with expression of CD8 [156].
Serial activation of TCRs by p–MHC ligation may elevate reactivity to low-affinity antigens and also help to discriminate between different high-affinity ligands. For example, it is important to discriminate between an acute infection that usually results in expression of high-affinity antigens at high density and a normal high-affinity antigen that escaped thymus presentation, which is often expressed at low density. Comparison of TCRs with different affinities showed that a TCR with an affinity greater than the physiologic range mediated stronger and faster responses than wild-type TCR. Paradoxically, this leads to an inability to recognize such an antigen presented in low density on target cells [157]. Ligation of high-affinity p–MHC with TCR can be relatively stable and impede ligation of this p–MHC with other TCRs. In this case, p–MHC in high density still may provide strong serial triggering. However, serial triggering should be blocked if p–MHC is presented in low density. In this fashion, TCR affinity and clustering can be considered part of peripheral immune tolerance.
The initial TCR/p–MHC binding is sterically limited because the exodomains of the TCR and p–MHC are short (~7 nm) requiring a relatively small intracellular cleft of ~15 nm to make contact [151]. This interaction is difficult because in the cell membrane the TCR neighbors highly abundant large surface proteins like CD45, whose extracellular segment ranges from 20 to 50 nm depending on the isoform [158]. Individual TCR/p–MHC interactions are short-lived (seconds), but for T cell activation, binding should continue for minutes to hours [151]. Therefore, a close contact between the T cell and target cell within a cleft of ~15 nm must be built and persist by exclusion of large surface moieties.
T cells use microvilli to create a close contact with the interrogated antigen-presenting cells (APCs) and target cells [24, 159]. These ~150 nm-diameter cell membrane protrusions contain adhesive receptors and form contact with target membranes with a short ~15 nm cleft, which sterically excludes CD45 and other bulky membrane proteins [151]. Microvilli usually last for seconds, but if a TCR complex presented in it binds agonist p–MHC, this can lead to cytoskeleton rearrangement that stabilizes the microvilli for a longer period of time [160]. The effective binding involves redistribution of adhesion receptors CD2 and LFA-1 that bind CD58 and ICAM-1 target proteins, respectively. The relatively long LFA-1/ICAM-1 interaction (~40 nm) moves to the periphery of the contact zone, while the shorter CD2/CD58 interaction (~15 nm) is placed in close proximity to the TCR/p–MHC binding [161].
The synapse is a dynamic structure that may contain various numbers of TCRs. Often it undergoes a transition from nanoclusters (~20) of TCR to microclusters (~300). These can proceed further to increased concentric membrane aggregates that in some conditions comprise up to 20% of the cell surface. Such complexes contain three concentric supramolecular activation clusters (SMACs): a central TCR/p–MHC cluster (cSMAC) with a narrow ~15 nm cleft [137], a peripheral ring of LFA-1/ICAM-1 (pSMAC) with a ~40 nm cleft, and a distal ring that includes CD45 and F-actin (dSMAC) in a bigger cleft [162]. This structure has been observed in helper, cytotoxic, and regulatory T cells [163].
The SMAC complex is a dynamic structure that can persist over a period of hours and provides signaling that can change in a timely fashion [16]. While cSMAC was initially considered only an activation domain, the recent identification of nearby late endosomal compartments suggests it can also function as a domain of TCR downregulation [161].
After completion of the immune response, T cell activation caused by TCR ligation should be downregulated. This can occur simply by exhaustion of available antigens, by negative feedback with synapse-associated cytosolic enzymes, or by specific checkpoint inhibitory receptors, such as PD-1, CTLA-4, B7-H3, DGK-α, LAG-3, and Tim-3, that are activated in parallel with the TCR and can work in the synapse by binding cognate target cell ligands [164]. PD-1 has two ligands, PD-L1 and PD-L2, which are members of the B7 family [165]. PD-1 is located in the immune synapse interface and recruits cytosol phosphatase SHP-2 to dephosphorylate CD28 [166]. PD-1 may also dephosphorylate phosphotyrosines in other TCR-associated proteins, such as Zap-70 and CD3 [165]. CTLA-4 shares two ligands, CD80 and CD86, with the stimulatory receptor, CD28, and can downregulate CD28 activity by binding its ligands [167, 168].
3.5. Signal Transduction Downstream of the p–MHC
Not only T cells but also target cells (“true targets” and APCs) can sense the immune synapse and react accordingly. For professional APCs, TCR/self-p–MHC contact during routine immune monitoring may cause signaling on the APC side to prevent its killing. After T cell attachment, DCs activate signaling mechanisms that facilitate cell–cell communication and actin and membrane remodeling [24, 169].
T cells can activate APCs by depositing their membrane fragments that contain TCRs, costimulatory and adhesion molecules, and cytokines on cognate antigen-bearing APCs. CD4+ T cells transfer to DCs membrane proteins by trogocytosis and the budding of T cell microvilli particles (TMP). The TMPs contain CD2, CD28, CD4, CD25, and activating cytokines and, upon uptake, initiate DC activation, including a calcium response and expression of costimulatory proteins such as CD40, CD80, and CD86 [24]. Memory CD8+ T cells release the DC-activating factor TNF-α, which induces the expression of an endogenous granzyme B inhibitor, PI-9, that protects DCs from killing by CD8+ effector T cells [170].
The reactions developed by cancer cells in response to T cell binding may include various pathways, such as induction of inhibitory ligands and cytokines [171, 172], abnormal tumor angiogenesis [173], downregulation of MHC expression [174], secretion of inhibitory exosomes [175], and complex modulation of the tumor microenvironment [176].
3.6. αβ TCR Clustering
Clustering and spatial cooperation of proteins in membranes are observed in many signaling pathways. Although TCR clustering may exist without antigen activation [100], such activation leads to a spatial reorganization of TCRs into signaling-competent clusters. In turn, initial TCR clustering may cooperatively facilitate further cluster development attracting additional resources from both membrane and cytosolic compartments [177].
The efficiency of the T cell reaction can be achieved either if the TCR has a relatively high affinity to p–MHC or if the affinity is relatively low, by cooperative involvement of neighboring TCRs. TCRs on the T cell membrane are usually localized in lipid rafts as 2D nanocluster aggregates, and the reaction on a single TCR may be amplified by lateral (horizontal) activation extension in the cluster. Therefore, structural rearrangement of activated TCR adaptors in the underlying cytosol might facilitate similar processes in its vicinity and promote cooperative lateral activation without additional p–MHC ligation. A strong positive cooperativity between individual TCRs has been detected in nanoclusters containing up to 20 TCRs [90, 178, 179]. In these experiments, binding of only two p–MHC (a p–MHC dimer) could stabilize 20 TCRs in the signaling-competent state [180]. Such allosteric reactions between TCRs can be an important factor of TCR selectivity that allows the detection of a relatively rare and weak “signal” (foreign p–MHC l) in the presence of abundant “noise” (self-p–MHC) [181]. Clustering of TCR complexes can be mediated by extracellular domain oligomerization, intracellular domain interactions, and attached cytoplasmic scaffold proteins [100].
In contrast to naïve T cells, TEFF and TMEM cells have a lower threshold of activation and response and also an elevated level of TCR clustering [178]. Unlike B cells, T cells lack the capacity to undergo “affinity maturation” after antigen engagement. However, “functional avidity maturation” can be achieved by TCR clustering [178, 182].
3.7. Structure and Signaling of the γδ TCR
The structure and function of the TCR in γδ T cells appear to be fairly different from αβ T cells. While the γδ TCR sometimes contains the same CD3 complex as the αβ TCR, for the recognition of homodimer, it uses TCR-γ and TCR-δ chains instead of TCR-α and TCR-β chains. In addition, Hayes et al. report that some γδ TCRs lack the CD3 δ chain and instead have the stoichiometry: γδ, γε, γε, and ζζ [183–186]. Similar to αβ TCRs, γδ TCRs are contained in lipid rafts and can form clusters on the cell surface [187]. Distinct from αβ T cells, γδ T cells do not require “classic MHC” molecules to recognize antigens, and they do not require CD4 or CD8 coreceptors.
Given the underlying differences, it is not surprising that signaling mechanisms in γδ T cells are distinct from αβ T cells. The γδ TCR has a stronger signaling capacity, which may be due to the fact that they constitutively express approximately twofold more of the TCR/CD3 complex than αβ T cells [188–190]. Also, the γδ TCR may provide signal transduction without a conformation shift of the CD3 complex. This quite unexpected divergence from the αβ TCR may be caused by differences in the TCR-γ and TCR-δ amino acid content, their glycosylation and orientation in the membrane, the pattern of TCR clustering, or the complement of associated kinases [191, 192]. For example, B lymphoid kinase (Blk), an Src family kinase expressed primarily in B cells, is expressed in γδ T cells but not in αβ T cells [193]. In addition, a subpopulation of γδ T cells, but not αβ T cells, has been detected in Lck-deficient and Zap-70-deficient mice, suggesting that Lck and Zap-70 are necessary for αβ T cell viability but not for γδ T cells [192]. In primary murine γδ T cells, TCRs contain ζζ homodimers. However, following ex vivo activation and expansion, one or both ζ subunits are replaced with FcεR1γ proteins [183].
4. Synthetic Receptors
4.1. Engineered TCRs
Synthetic TCRs (synTCRs) can potentially recognize all peptides processed and presented in the context of MHC molecules, thus allowing TCRs to target both surface and intracellular antigens. On the other hand, TCRs only recognize peptides in the context of the MHC complex. Therefore, this approach is hindered by some factors, including the need for MHC matching, MHC downregulation by cancer cells, suppressive tumor environment, and off-target/off-tumor killing [194]. In addition, the activity of TCR-transduced cells may be affected by the formation of mixed dimers between exogenous and endogenous α and β proteins which may decrease activity or lead to nonspecific reactivity [195, 196]. To prevent such dimerization, the selective binding between α and β proteins in synTCRs can be achieved by rearranging specifically interacting amino acid sequences in α and β constant domain interface [197] or by adding a second disulfide bond [198]. Another approach is to construct synTCRs with a murine constant region in place of the human constant region. This resulted in preferential pairing of the murine constant domains in α and β subunits and higher expression of the engineered TCR on the surface of the human lymphocytes [199]. One theoretical drawback in the use of mouse TCR domains might be the development of human anti-mouse TCR immune responses; however, this was not observed in clinical trials [200]. Expression of cancer-reactive γδ TCRs in αβ T cells prevents formation of mixed dimers [147]. This reprogramming accompanied by CRISPR-mediated elimination of the endogenous αβ TCR led to increased γδ TCR expression and efficiency irrespective of patient MHC type [201].
Besides optimization of amino acid content, several other factors should be considered in constructing a synthetic TCR. Adequate expression of exogenous αβ heterodimer depends on the configuration of the vector and expression cassettes. It also depends on availability of CD3 proteins (ε, ζ, δ, γ) for correct assembly in the Golgi [195]. Co-transfer of CD3 and αβ genes into primary murine T cells enhanced TCR expression and antigen-specific T cell function in vitro and in vivo [202]. TCRs with high affinity, not available in normal T cells because of negative selection in the thymus, may enhance the ability to kill target cells in the cancer environment [203]. Engineered T cells containing “high-affinity” TCRs showed efficiency in treating myeloma and synovial cell sarcomas [204, 205]. Also, generation of synthetic TCRs able to recognize specific cancer neo-antigens or known cancer-specific antigen/MHC combinations can be advantageous for developing individualized anticancer therapy [206, 207].
Another approach is to create synTCRs with antibody recognition domains. In these receptors, exodomains of α and β subunits of the TCR are modified by replacing their variable domains with antibody domains that can recognize cancer-associated antigens. It can be just variable domains, VH and VL [208], or Fab fragments with VH–CH domains fused over the TCR-α constant domain and VL–CL domains fused over the TCR-β constant domain [209]. The resulting chimeric TCR is expressed on the surface of cytotoxic T lymphocytes, recognizes antigen in a non-MHC-restricted manner, and transmits the signal through the CD3 complex for T cell activation [208]. The absence of p–MHC in the synapse excludes CD8 co-signaling; however, the affinity of Fab can be increased to a level that sufficiently compensates for the absence of CD8 [128].
4.2. Chimeric Antigen Receptors (CARs)
Basic chimeric antigen receptors (CARs) contains three elements: (1) a recognition domain that is a surface ligand-binding domain; (2) a transmembrane (TM) domain that is a structure ~20 amino acid long, enriched with hydrophobic amino acids (AAs) and forming an alpha helix in the cell membrane; and (3) an intracellular effector part that can contain various signaling domains needed for sustained effector cell ability to kill and propagate [208, 210].
Upon ligation to cognate antigens on the target cell, it is thought that the CAR dimerizes at the site of recognition and undergoes a conformational shift in its cytoplasmic domains, which leads to their phosphorylation, binding, and activation of Zap-70 with sequential activation of multiple signaling cascades [211]. In support of dimerization occurring, a mutation in a CAR’s ectodomain that facilitates spontaneous dimerization (even without antigen recognition) increased its functionality [212], whereas mutations in a CAR transmembrane domain that resulted in disruption of dimerization led to decreased CAR-T cell activation and cytolytic activity [213].
First-generation CARs contain a single-strand antibody (ScFv), TM, and CD3-ζ signaling domain [210, 214]. Second-generation CARs include a coactivator cytoplasmic domain in cis to provide additional T cell co-stimulation. The most widespread is a CD28 or 4–1BB signaling domain inserted between the TM and ζ domains. CD28 signals through activation of LCK, PI3K-Akt [215], Grb2, and Gads [216] and induces Bcl-XL [217] and IL2 [216]. 4–1BB signaling upon aggregation (trimerization) of 4–1BB ligand attracts TNF receptor-associated factors and forms a “signalosome” that activates T cell proliferation and survival [218]. This leads to phosphorylation of CD3 proteins ε and ζ, Lck, and LAT [219]. Other costimulatory domains, like ICOS, OX40, and CD27, can also function in CARs between the TM and ζ domains [220–222]. Third-generation CARs include two costimulatory domains, like CD28 and 4–1BB inserted between TM and ζ [223]. This additional co-stimulation apparently increases the basal activity of CARs and can be counterproductive due to baseline activation and auto-toxicity [224, 225].
CARs can apparently function in many different cytotoxic immunocytes [1, 226]. For human CD8+ T cells, the granzyme–perforin pathway seems to be the most common activated by the CAR, as this is the predominant cytotoxic mechanism in human T cells [46, 47, 227]. However, other pathways are also used as Hong et al. demonstrated Fas-mediated killing by CD30 CAR-T cells [228]. Because some CD4+ T cells possess cytotoxic activity, they also can be reprogrammed for CAR-mediated killing [227, 229]. Beyond conventional αβ T cells, CAR-mediated killing has also been shown in NK cells [230, 231], γδ T cells [232, 233], NKT cells [234, 235], and neutrophils [236]. While the mechanisms of killing by other effector cells reprogrammed with CARs might be more diverse, it is assumed that upon target recognition, CARs can activate the natural cytotoxic signaling pathways present in a host cell. Interestingly, for macrophages, a CAR that contains the cytosolic domains of Fc receptor instead of the ζ-signaling domain leads to phagocytosis upon target recognition instead of cytotoxicity [5].
In T cells, analysis of CAR-mediated targeting showed that affinity to cognate antigen in the interval of 10 μM to 1 mM allows for both effective recognition and dissociation when the T cell action is completed [237, 238]. However, lower affinity might be preferable to prevent off-tumor killing [239]. Steric hindrance both inside and outside the cell should be taken into consideration when designing a CAR. The length of the extracellular segment should be comparable with the optimal TCR/p–MHC distance at ~15 nm (see Subheading 3.4) [212, 240]. Likewise, steric limitations should be applicable to the cytosolic part of the CAR, because the signaling adaptor proteins should act in a certain distance from the cell membrane. Of course, the sophisticated TCR architecture makes it difficult to easily deduce the ideal CAR sequence. That is why during CAR construction, combinations of ecto-, TM, and endodomain amino acid contents have to be tested in parallel to determine variations in target affinity and signaling. It is important to note that even small differences in amino acid content can dramatically change the tertiary structure of the CAR, with obvious consequences for protein stability and function [241].
4.3. Comparing CARs with αβ TCRs
In engineering T cells, the simplicity of a CAR compared to a synTCR leads to both advantages and disadvantages. One advantage is that CARs recognize and bind targets independently of MHC and coreceptors like CD4 or CD8. In addition, CARs can recognize a wide spectrum of ligands on the cell surface, including proteins, carbohydrates [242], glycolipids, and other moieties [243], that are usually not recognized by TCRs. The CAR recognition motif structure appears flexible as scFvs, ligands (e.g., CD70 that binds CD27 receptor) [244], and single-strand avidin (that binds biotinilated targets) [245, 246] are all functional. Compared to TCRs, CARs provide a faster killing dynamic [149] and can be used in the presence of other CARs and TCRs in the host, both independently of them and in cooperation [247].
The main disadvantages of CARs are that they cannot target intracellular antigens and they do not communicate through the balanced system of CD3 proteins and coreceptors leading to less regulation in cytosolic signaling. That can undermine some important functions such as antigen recognition proofreading and adequate dynamics of the cell response. For example, first-generation CARs with a sole ζ chain as a signaling domain and without a coactivator signal were unable to maintain robust T cell viability in the presence of cognate antigen. In contrast to the TCR, the conventional CAR is not involved in cell monitoring and combinatorial antigen evaluation. Rather, it works as a binary operator that turns on the response as soon as it recognizes the target. Compared to conventional CTLs, CAR-T cells are less sensitive to p–MHC density. Whereas a CTL’s response may need only a few agonist p–MHCs per target cell [148–151], a CAR-T cell response may need about ~200 antigens per cell [248, 249].
Another major flaw of CARs compared to TCRs is that they usually produce a high basal signal, which can be deleterious for T cell viability [250]. Whereas the TCR emerges from the cell membrane as a tightly cooperative complex with accurate regulation of its subunits’ conformation, the CAR emerges as a single protein prone to specific and nonspecific reactions with surrounding molecules. Thus, while TCR tonic signal is caused by ligation with self-p–MHC and is a part of T cell homeostasis, the basal signal activity of the CAR is independent of antigen presentation and may disrupt T cell homeostasis. This basal activity is correlated with the density of CAR proteins on the cell membrane [250, 251]. In fact, CAR proteins can spontaneously aggregate in the cell membrane independent of external ligands, potentially because of thermodynamic driving factors and variation in physicochemical properties of CARs and surrounding proteins [252]. Multiple factors may determine CAR aggregation and toxicity including the spacer connecting the CAR’s recognition and TM domains [253], the configuration of the CAR’s active ITAMs [254], and the activity of T cell death signaling pathways, Fas and DR5 [255].
The comparison of CD19 CARs containing 4–1BB-ζ and CD28-ζ cytoplasmic tails showed that CD28-ζ CAR had higher spatial aggregation and more “basal” ζ phosphorylation and was more toxic for T cells [250]. However, basal CAR toxicity also has been shown for 4–1BB-ζ CAR [251].
4.4. Complications in the Clinical Use of CAR-T Therapy
Although CAR-T cells have been very successful in some clinical trials, this therapy is associated with serious complications. Among other factors, problems associated with CAR-T cells include uncontrollable activation, expansion, and persistence, as well as on-target/off-tumor and off-target/off-tumor killing. Upon introduction in patients, CART-19 cells can achieve rapid proliferation (up to 104-fold expansion), which may result in tumor lysis syndrome (TLS), cytokine release syndrome (CRS), and neurotoxicity [256–259]. In addition, CART-19 therapy kills all CD19+ cells leading to B cell aplasia [256]. On the other hand, B cell tumors with a mutated CD19 can escape CART-19 killing [260–262].
5. Next-Generation Strategies for T Cell Engineering
This quickly developing field has extended in many directions in an attempt to improve upon the original CAR and TCR approaches. Designs to improve both safety and efficacy include modifications of the CAR itself, combining multiple CARs, and adding multiple factors in addition to the CAR.
5.1. Construction of CARs with New Domains
Since the first description of a CAR, an impressive number of structural modifications with a wide number of variations have been introduced. Considerable attention has been focused on the discovery of novel scFv recognition domains to target different antigens. However, any structure able to bind a cancer cell is a theoretically viable alternative.
Several “universal” CAR systems have been constructed. In one strategy, the antigen-recognizing domain is replaced with a monomeric avidin moiety that binds biotin. Biotinylated tumor-specific molecules, such as a monoclonal antibody, can then direct the CAR-T cell to different target cells. Simply changing the biotinylated antibody redirects the CAR-T cell to recognize and trigger killing of cancer cells that are “stained” with the biotinylated monoclonal antibody [245, 246]. In a second strategy, Cho et al. replaced the scFv with a leucine zipper domain, such that the CAR could bind to a second chimeric protein composed of the cognate leucine interaction domain fused to an scFv. Introduction of such second chimeric proteins can continuously redirect the CAR-T cell activity [263]. A third strategy fuses a tumor antigen-specific Fab with a peptide that binds to a CAR. Peptide-associated Fabs that can have different specificities can be systemically delivered in vivo to connect the CAR-T with cognate antigen on the target cells. In this system, the antigenic diversity of Fabs and their dose determine CAR-T antigen specificity and the level of activation [212].
In addition to universal systems, another approach attempts to develop a CAR that can emulate the recognition ability of a TCR for agonist p–MHC. Here, a two-gene system is used. The first gene encodes a CAR where the scFv is replaced with a TCR-α chain that is truncated at the TM region and contains a cysteine [264]. The second gene encodes a TCR-β chain that is also truncated at the TM region and contains a cysteine to mediate disulfide bridging. When expressed, the TCR-β chain binds to the TCR-α chain of CAR, creating an αβ TCR/CAR hybrid. This approach increases the spectrum of antigen recognition, by inclusion of intracellular antigens, although in the context of MHC. It can be especially important in the treatment of cancer cells with neo-antigen markers as an alternative to TCR-mediated therapy.
Whereas the previous approach attempts to bring together TCR binding with CAR signaling, others have tried to create CARs with scFv binding that directly engage with TCR signaling. One approach to directly utilize TCR signaling is a “T cell antigen coupler” (TAC) [265]. In this design, the CAR contains an anticancer scFv recognition domain attached to the CD4 TM and signaling domains. In addition, a CD3-ε-binding domain is inserted in the CAR between the scFv and the TM, which results in CAR attachment to the CD3-ε subunit of the TCR. When the CAR’s scFv binds cognate antigen, the signal transduction goes through the CD4–TCR complex. In mouse models, this approach yielded increased antitumor efficacy with reduced toxicity. A second approach, called T cell receptor fusion constructs (TRuCs), was developed by sequentially attaching scFv domains to each subunit of the TCR [266]. In TRuC-T cells, the scFv is incorporated into the TCR and binding an antigen in an MHC-independent manner engages the signaling capacity of the entire TCR. Among the different TRuCs tested, fusing the scFv to the ε subunit showed the highest level of functionality. This is potentially due to its stoichiometric advantage as well as its specific cytoplasmic docking sites for Nck [116], GRK2 [267], CAST [268], and phospholipid-binding motif [269].
A separate tactic to increase CAR-T cell functionality is to insert in the CAR’s cytoplasmic tail domains that can emulate the signaling by homeostatic interleukins. Kagoya et al. introduced binding motifs for STAT5 and STAT3 in CD19 CAR and showed that the new CAR strengthened the activation of JAK kinase and STAT3 and STAT5 signals, elevated in vivo persistence of CAR-T cells, and increased their antitumor activity [270].
Finally, significant improvements can be made even without modifying any of the functional domains but only taking into account the tertiary structure of CAR protein and the distances between recognition domains and signaling domains that could be crucial for signaling. Using CD19 CAR, Ying et al. modified nonenzymatic “scaffold segments” of the CAR by increasing the length of the hinge and the distance between TM and signaling domains. By computer modeling, they showed that such variations can dramatically change tertiary structure of the protein. Then, by adding additional amino acids to the hinge and two amino acids between the TM and CD28-ζ, they were able to decrease the level of CAR signaling and the rate of CAR-T propagation in the presence of targets; and by that means virtually eliminate the development of chemokine shock in a mouse model and a clinical trial [241].
5.2. Combining More Than One CAR in a Cell
Expression of multiple constructs in T cells potentially can provide recognition of multiple antigens on the targets to discriminate between cancer cells and normal cells, and obtain adequate T cell signaling by switching both activation and inhibition pathways [221, 271–273].
This approach may increase CAR-T cell efficiency in situations where tumors downregulate the expression of cognate antigens. For example, failure of the CD19 CAR against B cell tumors is sometimes caused by internal deletions in CD19, which remove the recognized epitope [274]. In such a situation, the expression of an additional CAR that recognizes other B cell markers can prevent tumor escape [275]. This has been accomplished both by supplying two separate CARs and by supplying one CAR that contains two tandem scFvs, each for a different antigen [276]. In the latter case, because different antigens may have different length, the CAR should have appropriate sterical configuration of its recognition domains that not only allows simultaneous ligation with antigens but also maintains an appropriate distance to the target membrane for synapse formation.
A two-CAR system also may increase CAR-T cell selectivity. For example, consider the scenario where a normal cell has only one of two surface antigens present on a cancer cell. A T cell expressing two CARs, one that recognizes each antigen with sufficiently low affinity, may proceed with killing only when both CARs are simultaneously ligated with the target [273, 277]. In another scenario, a cancer cell may have only one of two surface antigens present on a normal cell. In this case, one CAR can recognize the shared antigen promoting killing, while a second CAR, known as an inhibitory CAR (iCAR), can recognize only normal cells and contain a signal inhibitory domain instead of the activation ζ domain. Fedorov et al. have shown that an iCAR with a segment of the inhibitory PD-1 cytoplasmic tail can prevent the stimulation of the other CARs when the T cell interacts with a cell containing both antigens [278].
Playing off the theme of converting positive to negative signals, switch receptors were designed to convert a negative signal to a positive one. Here, the extracellular domain of PD-1 is combined with the cytoplasmic domain of CD28. When the switch CAR binds the inhibitory ligand PD-L1, it activates T cells through the CD28-mediated pathway. In solid tumor models, T cells reprogrammed with both CAR and the switch receptor showed augmented efficacy compared to CAR-T cells [279–282].
5.3. Using Additional Genes to Reprogram the T Cells
The fourth generation of CARs, armored CARs, has been made by combining two expression cassettes: one coding a second-generation CAR and another coding an additional metabolically active protein, such as a cytokine, antibody, or another ligand [283]. Since CAR-T cells accumulate in the tumor, the active proteins are delivered locally to the site of disease, minimizing the toxicities often associated with active proteins delivered systemically. T cells loaded with a CAR construct armored with the proinflammatory cytokine IL12 showed elevated antitumor efficacy [284, 285]. CAR-T cells armored with IL18 increased CAR-T cell survival and enhanced immune response by modulating tumor microenvironment [286]. A CD20 CAR-T cell armored with IL7 and the chemokine ligand CCL19 improved immune cell infiltration and CAR-T cell survival in the tumor [287]. CAR-T cell persistence and efficiency can also be enhanced when cells are reprogrammed with constitutively active homeostatic receptors, such as IL2, IL7, or IL15 receptors, or with chimeric cytokine receptors that switch a negative signal produced by inhibitory cytokines, such as IL4, to a positive signal [288]. CD19 CAR-T cells, armored with the immune activator protein CD40L, exhibited increased cytotoxicity against CD40+ tumors and extended the survival of tumor-bearing mice in a xenotransplant model of CD19+ systemic lymphoma [289]. CAR-T cells loaded with a PD-1-blocking scFv enhanced the survival of PD-L1+ tumor-bearing mice in syngeneic and xenogeneic mouse models through both autocrine and paracrine mechanisms [290].
Suicide switch constructs can eliminate CAR-T cells upon systemic delivery of a signaling molecule. For example, this construct may contain caspase-9 protein fused with a protein that can be dimerized by a drug. In the presence of the drug, the caspase-9 dimerizes and activates the intrinsic apoptotic pathway [291]. A second strategy introduces a truncated epidermal growth factor receptor (EGFRt) that is recognized by the antibody cetuximab, which mediates antibody-dependent cellular cytotoxicity (ADCC) against the CAR-T cells [82]. However, kill switches do not allow control of the rate of T cell activation and expansion. Rather, they are turned on after the recognition of a problem and once activated cannot be reversed. In addition, leaky expression of an inducible suicide gene can undermine efficacy, while incomplete activation or targeting can undermine the purpose of the suicide switch.
Lim et al. improved the functionality of CAR-T cells by adding a synthetic Notch (synNotch) receptor that contains an antigen recognition domain (scFv), fused to the Notch regulatory core domain and a transcription activator domain. Binding of the cognate antigen stimulates cleavage of the receptor and releases the transcriptional activator, which can enter the nucleus and drive ectopic expression of genes inserted in the T cell DNA under an activator-specific promoter. One synNotch receptor, upon antigen binding, can activate multiple genes regulated by the same promoter [271, 292].
6. Challenges and Potential Future Solutions
A combinatorial approach for cancer treatment is important to emulate the complexity of the immune system in fighting tumors. Here, we will consider ways to (1) better emulate the TCR, (2) use other cells instead of αβ T cells, and (3) use other cells in combination with αβ T cells.
6.1. Better Approximation of the TCR
Conceptually, CAR-mediated activity may emulate TCR function with similar intercellular synapses and signaling pathways. Further improvement of CAR functionality might lay in better understanding the relationship between CARs and TCRs. CAR-mediated synapses are structurally and functionally different from TCR synapses, being smaller, less structured, and shorter-lived [149]. The CAR-T synapse, created without a TCR, is obviously deficient in coreceptors (CD8 or CD4) and in underlying processes related to coreceptor binding to MHC such as delivering additional Lck activator for Zap-70 (Subheading 3.3). In addition, they do not bind to MHC on target cells, so the target cell membrane state also can be different.
An important goal is to attain CARs that build a cooperative complex with CD3 (see Subheading 5.1). If the cytoplasmic tail of the CAR is not included in a tight TCR complex, it may constantly be in a “loose” conformation in the cytosol, prone to some degree of nonspecific aggregation or phosphorylation by Lck and basal signaling.
Optimizing the spatial and functional relationship of the CAR to the TCR is predicted to boost activity. Current experimental data suggests that the CAR–TCR relation can be quite flexible. T cells with defective TCR expression are still able to provide CAR-mediated killing [263, 293]. On the other hand, cells with an active TCR may contain TCRs as part of the CAR synapse [266]. A more integrated relationship between CARs and TCRs may be advantageous for CAR-T cell efficiency.
6.2. Expanding Beyond αβ T Cells
Another direction comes out of the fact that TCRs and CARs can work not only in αβ T cells but also in other immune cells involved in the innate response. In such cells, ectopic TCR and CAR activity can be combined with innate modalities to create a more complex immune response. As already mentioned, expression of endogenous TCRs has been shown in various myeloid cells, including eosinophils [294], neutrophils [79], monocytes, and macrophages [81, 294–296]. Ectopic TCRs and CARs that redirect immune cells against cognate targets can be expressed in NK cells [297, 298], NKT cells [299], γδ T cells, and cytokine-induced killer (CIK) cells [2]. In contrast to αβ T cells, certain subsets of innate immune cells have natural attraction to tumors through the recognition of stress-related tumor antigens.
γδ T cells serve as a particularly illustrative example. One reason they are attractive for ACT is that they have natural tropism for several types of cancer by monitoring stress-induced and inflammatory markers, such as lipopeptides, pyrophosphates, microorganism-derived proteins, and self-proteins through their γδ TCR, Toll-like, NK, and CD16 receptors [191, 192, 300]. In addition, they detect alterations in cell surface molecules, such as the MHC class I chain-related ligands A and B (MICA and MICB), and cell-associated antibodies [301, 302]. Their antitumor activity involves cytokine and chemokine secretion [303] and cytotoxicity. In contrast to αβ T cells, γδ T cells do not have strong lineage separation between helper and cytotoxic subsets, but do include TEFF, TCM, and TEM cells [303–307]. Cytotoxicity is accomplished through perforin–granzyme, TRAIL, FasL, and ADCC [308]. In addition to cytotoxicity, γδ T cells are able to phagocytose and present tumor antigens to CD8+ αβ Τ cells, as well as induce DC maturation by TNF-α secretion [309]. Finally, as an added advantage, γδ T cells are mainly not alloreactive and do not induce GVHD [310].
Both ectopic TCRs and CARs can be used to reprogram γδ T cells for adoptive therapy [301]. Transfer of ectopic αβ TCRs into γδ T cells can be provided without TCR mispairing and formation of mixed TCR heterodimers. γδ T cells engineered to express human αβ TCRs exhibited high levels of antitumor cytotoxic activity and cytokine release [311, 312]. Both GD2 and CD19 CARs have been used in peripheral blood-derived γδ T cells and shown to exhibit target-specific IFN-γ secretion and cytotoxicity [313, 314]. In the case of a GD2–CD28–CD3-ζ CAR, the reprogrammed γδ T cells showed enhanced GD2-specific killing beyond the γδ T cells without the CAR, which also recognized the tumor cells with endogenous “stress receptors.” Expanded CAR-T cells retained the ability to take up tumor antigens and cross present the processed peptide to responder αβ T cells [233]. To refine the specificity of γδ T cell cytotoxicity, Fisher et al. created a GD2 CAR in which the ζ domain had been replaced with innate NKG2D signaling molecule DAP10 [315]. These γδ CAR-T cells killed GD2+ glioblastoma cells, but not GD2+ control cells, by working with the endogenous γδ TCR targeting glioblastoma “stress receptors.” One way to augment the expression of tumor stress-related antigens can be by less specific treatments, like chemo- and radiotherapy [316].
Two obstacles to overcome in using γδ T cells are relatively low cell numbers, and they can also promote cancer progression by inhibiting antitumor responses, enhancing cancer angiogenesis, and increasing the population of myeloid-derived suppressor cells (MDSCs) [14, 317]. Thus, accurate amplification, evaluation, and selection of γδ T cell subsets will be an important part of their optimization for therapy. To some extent, the number and distinct activity of γδ T cells can be modulated with cytokine stimulation [14].
6.3. Total Immune System Engagement
The human immune system is very complex with various types of cells, proteins, and subcellular particles working synergistically against various diseases, including infection, cancer, and aging. The ability of pathological agents for fast propagation, modification, and population plasticity dictates the general immune system works as a multifactor combinatorial defense. Although today immune cell reprogramming is still limited to relatively simple combinations tested in clinical studies, it is clear that in the future, multi-cell, multi-ligand treatment will need to be developed for an efficient and flexible therapeutic approach. This can be especially important for the treatment of solid tumors that are often resistant to conventional CAR-T therapy [2] because of poor tumor recognition, penetration, and inhibition by the tumor microenvironment.
In addition to using T cells expressing multiple CARs and other chimeric receptors, B cells, NK cells, and other combinations of reprogrammed immune cells could be used. Most of the anticancer therapeutic designs are not mutually exclusive and can be applied in combinations to maximize the outcome. The improvement of therapy employing CARs and ectopic TCRs probably will depend on their cross talk with tumor-resident immunocytes. For example, the treatment may include co-introduction of αβ and γδ T cells reprogrammed with CARs and TCRs, DCs loaded ex vivo with tumor-specific antigens, cytokines, and checkpoint inhibitor ligands delivered locally to the tumors by “armored” T or NK cells. The versatile treatment will integrate the most efficacious combination of the helper, effector, and memory cells with their modulators that allow to maximize the therapeutic specificity and safety.
As a second example, tumor-infiltrating lymphocytes (TILs) are cells that can recognize and penetrate tumors. These cells can be extracted from the tumor, activated and propagated ex vivo, and then used for ACT. However, some solid tumors contain very low numbers of infiltrating T cells. So, combining therapies that increase T cell recruitment with subsequent TIL treatment might be beneficial. Although T cell recruitment can depend on chemokine gradients created by tumor-resident dendritic cells, the recruitment of DC in tumors is a process that is not fully understood. It may depend on multiple factors, including both activation and inhibition influenced by other tumor-resident cells such as macrophages and NK cells [318]. Here again, understanding better the mechanisms of intratumoral immune cell signaling might lead to approaches to more fully engage and reprogram multiple arms of the immune system.
7. Conclusions
T cell reprogramming has demonstrated that methods of genetic and cell engineering can be broadly used to great clinical benefit by augmenting the body’s own immune defense. Simple designs combining a few domains are starting to give way to more complicated approaches that either more closely emulate actual TCR signaling or integrate secondary signaling pathways. Similar approaches have been successfully used in reprogramming other immunocytes and have rather universal applicability in cell biology for other cell types and other diseases. This is especially encouraging for the development of combination therapies that employ multiple reprogramming factors and reprogrammed immune cells, with accurate monitoring of clinical outcome and fast adjustments of the treatment. The accumulation of knowledge from numerous academic, biopharmaceutical, and clinical results is rapidly translating our thought processes and ability to conquer cancer.
Acknowledgment
Sources of Support: NIH R21CA198561, NIH R21AI121993, and Alliance for Cancer Gene Therapy.
References
- 1.Harrer DC, Dorrie J, Schaft N (2018) Chimeric antigen receptors in different cell types: new vehicles join the race. Hum Gene Ther 29(5):547–558. 10.1089/hum.2017.236 [DOI] [PubMed] [Google Scholar]
- 2.Rotolo R, Leuci V, Donini C, Cykowska A, Gammaitoni L, Medico G, Valabrega G, Aglietta M, Sangiolo D (2019) CAR-based strategies beyond T lymphocytes: integrative opportunities for cancer adoptive immunotherapy. Int J Mol Sci 20(11):2839 10.3390/ijms20112839 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 3.Mehta RS, Rezvani K (2018) Chimeric antigen receptor expressing natural killer cells for the immunotherapy of cancer. Front Immunol 9:283 10.3389/fimmu.2018.00283 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 4.Cassetta L, Kitamura T (2018) Macrophage targeting: opening new possibilities for cancer immunotherapy. Immunology 155(3):285–293. 10.1111/imm.12976 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 5.Morrissey MA, Williamson AP, Steinbach AM, Roberts EW, Kern N, Headley MB, Vale RD (2018) Chimeric antigen receptors that trigger phagocytosis. elife 7:e36688 10.7554/eLife.36688 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 6.Jurgens B, Clarke NS (2019) Evolution of CAR T-cell immunotherapy in terms of patenting activity. Nat Biotechnol 37(4):370–375. 10.1038/s41587-019-0083-5 [DOI] [PubMed] [Google Scholar]
- 7.Boyiadzis MM, Dhodapkar MV, Brentjens RJ, Kochenderfer JN, Neelapu SS, Maus MV, Porter DL, Maloney DG, Grupp SA, Mackall CL, June CH, Bishop MR (2018) Chimeric antigen receptor (CAR) T therapies for the treatment of hematologic malignancies: clinical perspective and significance. J Immunother Cancer 6(1):137 10.1186/s40425-018-0460-5 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 8.EMA (2018) First two CAR-T cell medicines recommended for approval in the European Union. European Medicines Agency, Amsterdam [Google Scholar]
- 9.Di Rosa F, Pabst R (2005) The bone marrow: a nest for migratory memory T cells. Trends Immunol 26(7):360–366. 10.1016/j.it.2005.04.011 [DOI] [PubMed] [Google Scholar]
- 10.Dhodapkar MV, Kumar V (2017) Type II NKT cells and their emerging role in health and disease. J Immunol 198(3):1015–1021 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 11.Fan X, Rudensky AY (2016) Hallmarks of tissue-resident lymphocytes. Cell 164(6):1198–1211. 10.1016/j.cell.2016.02.048 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 12.Godfrey DI, Uldrich AP, McCluskey J, Rossjohn J, Moody DB (2015) The burgeoning family of unconventional T cells. Nat Immunol 16(11):1114–1123 [DOI] [PubMed] [Google Scholar]
- 13.Ziegler H, Welker C, Sterk M, Haarer J, Rammensee HG, Handgretinger R, Schilbach K (2014) Human peripheral CD4(+) Vdelta1(+) gammadeltaT cells can develop into alphabetaT cells. Front Immunol 5:645 10.3389/fimmu.2014.00645 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 14.Zou C, Zhao P, Xiao Z, Han X, Fu F, Fu L (2017) gammadelta T cells in cancer immunotherapy. Oncotarget 8(5):8900–8909. 10.18632/oncotarget.13051 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 15.Khairallah C, Chu TH, Sheridan BS (2018) Tissue adaptations of memory and tissue-resident gamma delta T cells. Front Immunol 9:2636 10.3389/fimmu.2018.02636 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 16.Alcover A, Alarcon B, Bartolo VD (2018) Cell biology of T cell receptor expression and regulation. Annu Rev Immunol 36:103–125. 10.1146/annurev-immunol-042617-053429 [DOI] [PubMed] [Google Scholar]
- 17.Stritesky GL, Jameson SC, Hogquist KA (2012) Selection of self-reactive T cells in the thymus. Annu Rev Immunol 30:95–114 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 18.Serroukh Y, Gu-Trantien C, Hooshiar Kashani B, Defrance M, Vu Manh TP, Azouz A, Detavernier A, Hoyois A, Das J, Bizet M, Pollet E, Tabbuso T, Calonne E, van Gisbergen K, Dalod M, Fuks F, Goriely S, Marchant A (2018) The transcription factors Runx3 and ThPOK cross-regulate acquisition of cytotoxic function by human Th1 lymphocytes. Elife 7:e30496 10.7554/eLife.30496 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 19.Mucida D, Husain MM, Muroi S, van Wijk F, Shinnakasu R, Naoe Y, Reis BS, Huang Y, Lambolez F, Docherty M, Attinger A, Shui JW, Kim G, Lena CJ, Sakaguchi S, Miyamoto C, Wang P, Atarashi K, Park Y, Nakayama T, Honda K, Ellmeier W, Kronenberg M, Taniuchi I, Cheroutre H (2013) Transcriptional reprogramming of mature CD4(+) helper T cells generates distinct MHC class II-restricted cytotoxic T lymphocytes. Nat Immunol 14(3):281–289. 10.1038/ni.2523 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 20.Takeuchi A, Saito T (2017) CD4 CTL, a cytotoxic subset of CD4+ T cells, their differentiation and function. Front Immunol 8:194. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 21.Oja AE, Vieira Braga FA, Remmerswaal EBM, Kragten NAM, Hertoghs KML, Zuo J, Moss PA, van Lier RAW, van Gisbergen KPJM, Hombrink P (2017) The transcription factor Hobit identifies human cytotoxic CD4+ T cells. Front Immunol 8:325. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 22.Hombach A, Kohler H, Rappl G, Abken H (2006) Human CD4+ T cells lyse target cells via granzyme/perforin upon circumvention of MHC class II restriction by an antibody-like immunoreceptor. J Immunol 177(8):5668–5675 [DOI] [PubMed] [Google Scholar]
- 23.Raeber ME, Zurbuchen Y, Impellizzieri D, Boyman O (2018) The role of cytokines in T-cell memory in health and disease. Immunol Rev 283(1):176–193. 10.1111/imr.12644 [DOI] [PubMed] [Google Scholar]
- 24.Kim HR, Mun Y, Lee KS, Park YJ, Park JS, Park JH, Jeon BN, Kim CH, Jun Y, Hyun YM, Kim M, Lee SM, Park CS, Im SH, Jun CD (2018) T cell microvilli constitute immunological synaptosomes that carry messages to antigen-presenting cells. Nat Commun 9(1):3630 10.1038/s41467-018-06090-8 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 25.Myers DR, Zikherman J, Roose JP (2017) Tonic signals: why do lymphocytes bother? Trends Immunol 38(11):844–857. 10.1016/j.it.2017.06.010 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 26.Born WK, Kemal Aydintug M, O’Brien RL (2013) Diversity of gammadelta T-cell antigens. Cell Mol Immunol 10(1):13–20. 10.1038/cmi.2012.45 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 27.Qi Q, Xia M, Hu J, Hicks E, Iyer A, Xiong N, August A (2009) Enhanced development of CD4+ gammadelta T cells in the absence of Itk results in elevated IgE production. Blood 114(3):564–571. 10.1182/blood-2008-12-196345 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 28.Brownlie RJ, Zamoyska R (2013) T cell receptor signalling networks: branched, diversified and bounded. Nat Rev Immunol 13(4):257–269 [DOI] [PubMed] [Google Scholar]
- 29.Geltink RIK, Kyle RL, Pearce EL (2018) Unraveling the complex interplay between T cell metabolism and function. AnnuRev Immunol 36:461–488. 10.1146/annurev-immunol-042617-053019 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 30.Kershaw MH, Westwood JA, Darcy PK (2013) Gene-engineered T cells for cancer therapy. Nat Rev Cancer 13(8):525–541. 10.1038/nrc3565 [DOI] [PubMed] [Google Scholar]
- 31.Mescher MF, Curtsinger JM, Agarwal P, Casey KA, Gerner M, Hammerbeck CD, Popescu F, Xiao Z (2006) Signals required for programming effector and memory development by CD8+ T cells. Immunol Rev 211:81–92. 10.1111/j.0105-2896.2006.00382.x [DOI] [PubMed] [Google Scholar]
- 32.Kolumam GA, Thomas S, Thompson LJ, Sprent J, Murali-Krishna K (2005) Type I interferons act directly on CD8 T cells to allow clonal expansion and memory formation in response to viral infection. J Exp Med 202(5):637–650. 10.1084/jem.20050821 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 33.Wrzesinski C, Restifo NP (2005) Less is more: lymphodepletion followed by hematopoietic stem cell transplant augments adoptive T-cell-based anti-tumor immunotherapy. Curr Opin Immunol 17(2):195–201 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 34.Brenchley JM, Douek DC, Ambrozak DR, Chatterji M, Betts MR, Davis LS, Koup RA (2002) Expansion of activated human naive T-cells precedes effector function. Clin Exp Immunol 130(3):432–440. 10.1046/j.1365-2249.2002.02015.x [DOI] [PMC free article] [PubMed] [Google Scholar]
- 35.Jones RG, Pearce EJ (2017) MenTORing immunity: mTOR signaling in the development and function of tissue-resident immune cells. Immunity 46(5):730–742. 10.1016/j.immuni.2017.04.028 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 36.McLean AR, Michie CA (1995) In vivo estimates of division and death rates of human T lymphocytes. Proc Natl Acad Sci U S A 92(9):3707–3711. 10.1073/pnas.92.9.3707 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 37.Bains I, Antia R, Callard R, Yates AJ (2009) Quantifying the development of the peripheral naive CD4+ T-cell pool in humans. Blood 113(22):5480–5487. 10.1182/blood-2008-10-184184 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 38.De Boer RJ, Homann D, Perelson AS (2003) Different dynamics of CD4+ and CD8+ T cell responses during and after acute lymphocytic choriomeningitis virus infection. J Immunol 171(8):3928–3935 [DOI] [PubMed] [Google Scholar]
- 39.Yoon H, Kim TS, Braciale TJ (2010) The cell cycle time of CD8+ T cells responding in vivo is controlled by the type of antigenic stimulus. PLoS One 5(11):e15423. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 40.Arsenio J, Metz PJ, Chang JT (2015) Asymmetric cell division in T lymphocyte fate diversification. Trends Immunol 36(11):670–683. 10.1016/j.it.2015.09.004 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 41.Pollizzi KN, Sun I-H, Patel CH, Lo Y-C, Oh M-H, Waickman AT, Tam AJ, Blosser RL, Wen J, Delgoffe GM, Powell JD (2016) Asymmetric inheritance of mTORC1 kinase activity during division dictates CD8(+) T cell differentiation. Nat Immunol 17(6):704–711 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 42.van der Windt GJW, Everts B, Chang C-H, Curtis JD, Freitas TC, Amiel E, Pearce EJ, Pearce EL (2012) Mitochondrial respiratory capacity is a critical regulator of CD8+ T cell memory development. Immunity 36(1):68–78 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 43.Sukumar M, Liu J, Ji Y, Subramanian M, Crompton JG, Yu Z, Roychoudhuri R, Palmer DC, Muranski P, Karoly ED, Mohney RP, Klebanoff CA, Lal A, Finkel T, Restifo NP, Gattinoni L (2013) Inhibiting glycolytic metabolism enhances CD8+ T cell memory and antitumor function. J Clin Invest 123(10):4479–4488 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 44.Chapman NM, Chi H (2018) Hallmarks of T-cell exit from quiescence. Cancer Immunol Res 6(5):502–508. 10.1158/2326-6066.CIR-17-0605 [DOI] [PubMed] [Google Scholar]
- 45.Martinez-Lostao L, Anel A, Pardo J (2015) How do cytotoxic lymphocytes kill cancer cells? Clin Cancer Res 21(22):5047–5056. 10.1158/1078-0432.CCR-15-0685 [DOI] [PubMed] [Google Scholar]
- 46.Masaki Y, Ohminami H, Arai J, Kasahara Y, Ishida Y, Fujita S (2002) Granule exocytosis, and not the Fas/Fas ligand system, is the main pathway of cytotoxicity mediated by alloantigen-specific CD41 as well as CD81 cytotoxic T lymphocytes in humans. Blood 95(7):2352–2356 [PubMed] [Google Scholar]
- 47.Benmebarek MR, Karches CH, Cadilha BL, Lesch S, Endres S, Kobold S (2019) Killing mechanisms of chimeric antigen receptor (CAR) T cells. Int J Mol Sci 20(6):E1283 10.3390/ijms20061283 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 48.Cullen SP, Martin SJ (2008) Mechanisms of granule-dependent killing. Cell Death Differ 15(2):251–262. 10.1038/sj.cdd.4402244 [DOI] [PubMed] [Google Scholar]
- 49.de Saint Basile G, Menasche G, Fischer A (2010) Molecular mechanisms of biogenesis and exocytosis of cytotoxic granules. Nat Rev Immunol 10(8):568–579. 10.1038/nri2803 [DOI] [PubMed] [Google Scholar]
- 50.Zhan Y, Carrington EM, Zhang Y, Heinzel S, Lew AM (2017) Life and death of activated T cells: how are they different from naive T cells? Front Immunol 8:1809 10.3389/fimmu.2017.01809 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 51.Malyshkina A, Littwitz-Salomon E, Sutter K, Zelinskyy G, Windmann S, Schimmer S, Paschen A, Streeck H, Hasenkrug KJ, Dittmer U (2017) Fas Ligand-mediated cytotoxicity of CD4+ T cells during chronic retrovirus infection. Sci Rep 7(1):7785 10.1038/s41598-017-08578-7 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 52.Mirandola P, Ponti C, Gobbi G, Sponzilli I, Vaccarezza M, Cocco L, Zauli G, Secchiero P, Manzoli FA, Vitale M (2004) Activated human NK and CD8+ T cells express both TNF-related apoptosis-inducing ligand (TRAIL) and TRAIL receptors but are resistant to TRAIL-mediated cytotoxicity. Blood 104(8):2418–2424 [DOI] [PubMed] [Google Scholar]
- 53.Marrack P, Scott-Browne J, MacLeod MK (2010) Terminating the immune response. Immunol Rev 236:5–10. 10.1111/j.1600-065X.2010.00928.x [DOI] [PMC free article] [PubMed] [Google Scholar]
- 54.Sallusto F, Lenig D, Forster R, Lipp M, Lanzavecchia A (1999) Two subsets of memory T lymphocytes with distinct homing potentials and effector functions. Nature 401(6754):708–712 [DOI] [PubMed] [Google Scholar]
- 55.Masopust D, Vezys V, Marzo AL, Lefrancois L (2001) Preferential localization of effector memory cells in nonlymphoid tissue. Science 291(5512):2413–2417 [DOI] [PubMed] [Google Scholar]
- 56.Macallan DC, Wallace D, Zhang Y, De Lara C, Worth AT, Ghattas H, Griffin GE, Beverley PCL, Tough DF (2004) Rapid turnover of effector-memory CD4(+) T cells in healthy humans. J Exp Med 200(2):255–260 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 57.Amsen D, van Gisbergen KPJM, Hombrink P, van Lier RAW (2018) Tissue-resident memory T cells at the center of immunity to solid tumors. Nat Immunol 19(6):538–546 [DOI] [PubMed] [Google Scholar]
- 58.Farber DL, Yudanin NA, Restifo NP (2014) Human memory T cells: generation, compartmentalization and homeostasis. Nat Rev Immunol 14(1):24–35 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 59.Willinger T, Freeman T, Herbert M, Hasegawa H, McMichael AJ, Callan MF (2006) Human naive CD8 T cells down-regulate expression of the WNT pathway transcription factors lymphoid enhancer binding factor 1 and transcription factor 7 (T cell factor-1) following antigen encounter in vitro and in vivo. J Immunol 176(3):1439–1446. 10.4049/jimmunol.176.3.1439 [DOI] [PubMed] [Google Scholar]
- 60.Gattinoni L, Zhong XS, Palmer DC, Ji Y, Hinrichs CS, Yu Z, Wrzesinski C, Boni A, Cassard L, Garvin LM, Paulos CM, Muranski P, Restifo NP (2009) Wnt signaling arrests effector T cell differentiation and generates CD8+ memory stem cells. Nat Med 15(7):808–813. 10.1038/nm.1982 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 61.Gattinoni L, Lugli E, Ji Y, Pos Z, Paulos CM, Quigley MF, Almeida JR, Gostick E, Yu Z, Carpenito C, Wang E, Douek DC, Price DA, June CH, Marincola FM, Roederer M, Restifo NP (2011) A human memory T cell subset with stem cell-like properties. Nat Med 17(10):1290–1297. 10.1038/nm.2446 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 62.Youngblood B, Hale JS, Kissick HT, Ahn E, Xu X, Wieland A, Araki K, West EE, Ghoneim HE, Fan Y, Dogra P, Davis CW, Konieczny BT, Antia R, Cheng X, Ahmed R (2017) Effector CD8 T cells dedifferentiate into long-lived memory cells. Nature 552(7685):404–409 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 63.Schenkel JM, Masopust D (2014) Tissue-resident memory T cells. Immunity 41(6):886–897 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 64.Martin MD, Badovinac VP (2018) Defining memory CD8 T cell. Front Immunol 9:2692 10.3389/fimmu.2018.02692 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 65.Lugli E, Gattinoni L, Roberto A, Mavilio D, Price DA, Restifo NP, Roederer M (2013) Identification, isolation and in vitro expansion of human and nonhuman primate T stem cell memory cells. Nat Protoc 8(1):33–42 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 66.Kawabe T, Jankovic D, Kawabe S, Huang Y, Lee PH, Yamane H, Zhu J, Sher A, Germain RN, Paul WE (2017) Memory-phenotype CD4(+) T cells spontaneously generated under steady-state conditions exert innate TH1-like effector function. Sci Immunol 2(12). 10.1126/sciimmunol.aam9304 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 67.Demissie E, Mahajan VS, Alsufyani F, Kumari S, Yuen GJ, Viswanadham V, Tran JQ, Moon JJ, Irvine DJ, Pillai S (2019) DOCK2 sets the threshold for entry into the virtual memory CD8+ T cell compartment by negatively regulating tonic TCR triggering. bioRXiv. 10.1101/582486 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 68.White JT, Cross EW, Burchill MA, Danhorn T, McCarter MD, Rosen HR, O’Connor B, Kedl RM (2016) Virtual memory T cells develop and mediate bystander protective immunity in an IL-15-dependent manner. Nat Commun 7:11291. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 69.Anthony SM, Howard ME, Hailemichael Y, Overwijk WW, Schluns KS (2015) Soluble interleukin-15 complexes are generated in vivo by type I interferon dependent and independent pathways. PLoS One 10(3):e0120274. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 70.Geginat J, Sallusto F, Lanzavecchia A (2001) Cytokine-driven proliferation and differentiation of human naive, central memory, and effector memory CD4(+) T cells. J Exp Med 194(12):1711–1719 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 71.Westermann J, Bode U, Sahle A, Speck U, Karin N, Bell EB, Kalies K, Gebert A (2005) Naive, effector, and memory T lymphocytes efficiently scan dendritic cells in vivo: contact frequency in T cell zones of secondary lymphoid organs does not depend on LFA-1 expression and facilitates survival of effector T cells. J Immunol 174(5):2517–2524 [DOI] [PubMed] [Google Scholar]
- 72.Nolz JC, Rai D, Badovinac VP, Harty JT (2012) Division-linked generation of death-intermediates regulates the numerical stability of memory CD8 T cells. Proc Natl Acad Sci U S A 109(16):6199–6204 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 73.Vrisekoop N, den Braber I, de Boer AB, Ruiter AFC, Ackermans MT, van der Crabben SN, Schrijver EHR, Spierenburg G, Sauerwein HP, Hazenberg MD, de Boer RJ, Miedema F, Borghans JAM, Tesselaar K (2008) Sparse production but preferential incorporation of recently produced naive T cells in the human peripheral pool. Proc Natl Acad Sci U S A 105(16):6115–6120 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 74.Biasco L, Scala S, Basso Ricci L, Dionisio F, Baricordi C, Calabria A, Giannelli S, Cieri N, Barzaghi F, Pajno R, Al-Mousa H, Scarselli A, Cancrini C, Bordignon C, Roncarolo MG, Montini E, Bonini C, Aiuti A (2015) In vivo tracking of T cells in humans unveils decade-long survival and activity of genetically modified T memory stem cells. Sci Transl Med 7(273):273ra213. [DOI] [PubMed] [Google Scholar]
- 75.Oliveira G, Ruggiero E, Stanghellini MTL, Cieri N, D’Agostino M, D’Agostino M, Fronza R, Lulay C, Dionisio F, Mastaglio S, Greco R, Peccatori J, Aiuti A, Ambrosi A, Biasco L, Bondanza A, Lambiase A, Traversari C, Vago L, von Kalle C, Schmidt M, Bordignon C, Ciceri F, Bonini C (2015) Tracking genetically engineered lymphocytes long-term reveals the dynamics of T cell immunological memory. Sci Transl Med 7(317):317ra198. [DOI] [PubMed] [Google Scholar]
- 76.Fagnoni FF, Vescovini R, Passeri G, Bologna G, Pedrazzoni M, Lavagetto G, Casti A, Franceschi C, Passeri M, Sansoni P (2000) Shortage of circulating naive CD8(+) T cells provides new insights on immunodeficiency in aging. Blood 95(9):2860–2868 [PubMed] [Google Scholar]
- 77.Naylor K, Li G, Vallejo AN, Lee W-W, Koetz K, Bryl E, Witkowski J, Fulbright J, Weyand CM, Goronzy JJ (2005) The influence of age on T cell generation and TCR diversity. J Immunol 174(11):7446–7452 [DOI] [PubMed] [Google Scholar]
- 78.Yager EJ, Ahmed M, Lanzer K, Randall TD, Woodland DL, Blackman MA (2008) Age-associated decline in T cell repertoire diversity leads to holes in the repertoire and impaired immunity to influenza virus. J Exp Med 205(3):711–723 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 79.Puellmann K, Kaminski WE, Vogel M, Nebe CT, Schroeder J, Wolf H, Beham AW (2006) A variable immunoreceptor in a subpopulation of human neutrophils. Proc Natl Acad Sci U S A 103(39):14441–14446. 10.1073/pnas.0603406103 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 80.Fuchs T, Puellmann K, Scharfenstein O, Eichner R, Stobe E, Becker A, Pechlivanidou I, Kzhyshkowska J, Gratchev A, Ganser A, Neumaier M, Beham AW, Kaminski WE (2012) The neutrophil recombinatorial TCR-like immune receptor is expressed across the entire human life span but repertoire diversity declines in old age. Biochem Biophys Res Commun 419(2):309–315. 10.1016/j.bbrc.2012.02.017 [DOI] [PubMed] [Google Scholar]
- 81.Fuchs T, Puellmann K, Emmert A, Fleig J, Oniga S, Laird R, Heida NM, Schafer K, Neumaier M, Beham AW, Kaminski WE (2015) The macrophage-TCRalphabeta is a cholesterol-responsive combinatorial immune receptor and implicated in atherosclerosis. Biochem Biophys Res Commun 456(1):59–65. 10.1016/j.bbrc.2014.11.034 [DOI] [PubMed] [Google Scholar]
- 82.Kao RL, Truscott LC, Chiou TT, Tsai W, AM W, De Oliveira SN (2019) Cetuximab-mediated suicide system in chimeric antigen receptor-modified hematopoietic stem cells for cancer therapy. Hum Gene Ther 30(4):413–428 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 83.Brazin KN, Mallis RJ, Boeszoermenyi A, Feng Y, Yoshizawa A, Reche PA, Kaur P, Bi K, Hussey RE, Duke-Cohan JS, Song L, Wagner G, Arthanari H, Lang MJ, Reinherz EL (2018) The T cell antigen receptor alpha transmembrane domain coordinates triggering through regulation of Bilayer immersion and CD3 subunit associations. Immunity 49(5):829–841.e826. 10.1016/j.immuni.2018.09.007 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 84.Wucherpfennig KW, Gagnon E, Call MJ, Huseby ES, Call ME (2010) Structural biology of the T-cell receptor: insights into receptor assembly, ligand recognition, and initiation of signaling. Cold Spring Harb Perspect Biol 2(4):a005140 10.1101/cshperspect.a005140 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 85.Call ME, Wucherpfennig KW (2004) Molecular mechanisms for the assembly of the T cell receptor-CD3 complex. Mol Immunol 40(18):1295–1305. 10.1016/j.molimm.2003.11.017 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 86.Love PE, Hayes SM (2010) ITAM-mediated signaling by the T-cell antigen receptor. Cold Spring Harb Perspect Biol 2(6):a002485 10.1101/cshperspect.a002485 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 87.Park S, Krshnan L, Call MJ, Call ME, Im W (2018) Structural conservation and effects of alterations in T cell receptor transmembrane interfaces. Biophys J 114(5):1030–1035. 10.1016/j.bpj.2018.01.004 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 88.Deswal S, Schamel WWA (2012) CD3ζ In: Choi S (ed) Encyclopedia of signaling molecules. Springer, New York, pp 294–314. 10.1007/978-1-4419-0461-4 [DOI] [Google Scholar]
- 89.Rojo JM, Portoles P (1991) A symmetrical view of the T-cell receptor-CD3 complex. Immunol Today 12(10):377–378 [DOI] [PubMed] [Google Scholar]
- 90.Schamel WW, Alarcon B (2013) Organization of the resting TCR in nanoscale oligomers. Immunol Rev 251(1):13–20. 10.1111/imr.12019 [DOI] [PubMed] [Google Scholar]
- 91.Exley M, Wileman T, Mueller B, Terhorst C (1995) Evidence for multivalent structure of T-cell antigen receptor complex. Mol Immunol 32(11):829–839 [DOI] [PubMed] [Google Scholar]
- 92.Fernandez-Miguel G, Alarcon B, Iglesias A, Bluethmann H, Alvarez-Mon M, Sanz E, de la Hera A (1999) Multivalent structure of an alphabetaT cell receptor. Proc Natl Acad Sci U S A 96(4):1547–1552. 10.1073/pnas.96.4.1547 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 93.Balagopalan L, Sherman E, Barr VA, Samelson LE (2011) Imaging techniques for assaying lymphocyte activation in action. Nat Rev Immunol 11(1):21–33. 10.1038/nri2903 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 94.Lee K-H, Dinner AR, Tu C, Campi G, Raychaudhuri S, Varma R, Sims TN, Burack WR, Wu H, Wang J, Kanagawa O, Markiewicz M, Allen PM, Dustin ML, Chakraborty AK, Shaw AS (2003) The immunological synapse balances T cell receptor signaling and degradation. Science 302(5648):1218–1222 [DOI] [PubMed] [Google Scholar]
- 95.Eleftheriadis T, Kartsios C, Yiannaki E, Kazila P, Antoniadi G, Liakopoulos V, Markala D (2008) Chronic inflammation and CD16+ natural killer cell zeta-chain downregulation in hemodialysis patients. Blood Purif 26(4):317–321. 10.1159/000130068 [DOI] [PubMed] [Google Scholar]
- 96.Baudouin SJ, Angibaud J, Loussouarn G, Bonnamain V (2008) The signaling adaptor protein CD3ζ is a negative regulator dendrite development young neurons. Mol Biol Cell 19:2444–2456. 10.1091/mbc.E07-09-0947 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 97.Krshnan L, Park S, Im W, Call MJ, Call ME (2016) A conserved alphabeta transmembrane interface forms the core of a compact T-cell receptor-CD3 structure within the membrane. Proc Natl Acad Sci U S A 113(43):E6649–E6658. 10.1073/pnas.1611445113 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 98.Xu C, Gagnon E, Call ME, Schnell JR, Schwieters CD, Carman CV, Chou JJ, Wucherpfennig KW (2008) Regulation of T cell receptor activation by dynamic membrane binding of the CD3epsilon cytoplasmic tyrosine-based motif. Cell 135(4):702–713. 10.1016/j.cell.2008.09.044 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 99.Gagnon E, Schubert DA, Gordo S, Chu HH, Wucherpfennig KW (2012) Local changes in lipid environment of TCR microclusters regulate membrane binding by the CD3epsilon cytoplasmic domain. J Exp Med 209(13):2423–2439. 10.1084/jem.20120790 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 100.Wang JH, Reinherz EL (2012) The structural basis of alphabeta T-lineage immune recognition: TCR docking topologies, mechanotransduction, and co-receptor function. Immunol Rev 250(1):102–119. 10.1111/j.1600-065X.2012.01161.x [DOI] [PMC free article] [PubMed] [Google Scholar]
- 101.Minguet S, Schamel WW (2008) A permissive geometry model for TCR-CD3 activation. Trends Biochem Sci 33(2):51–57. 10.1016/j.tibs.2007.10.008 [DOI] [PubMed] [Google Scholar]
- 102.Zhang H, Cordoba SP, Dushek O, van der Merwe PA (2011) Basic residues in the T-cell receptor zeta cytoplasmic domain mediate membrane association and modulate signaling. Proc Natl Acad Sci U S A 108(48):19323–19328. 10.1073/pnas.1108052108 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 103.Paensuwan P, Hartl FA, Yousefi OS, Ngoenkam J, Wipa P, Beck-Garcia E, Dopfer EP, Khamsri B, Sanguansermsri D, Minguet S, Schamel WW, Pongcharoen S (2016) Nck binds to the T cell antigen receptor using its SH3.1 and SH2 domains in a cooperative manner, promoting TCR functioning. J Immunol 196(1):448–458 [DOI] [PubMed] [Google Scholar]
- 104.Holst J, Wang H, Eder KD, Workman CJ, Boyd KL, Baquet Z, Singh H, Forbes K, Chruscinski A, Smeyne R, van Oers NSC, Utz PJ, Vignali DAA (2008) Scalable signaling mediated by T cell antigen receptor-CD3 ITAMs ensures effective negative selection and prevents autoimmunity. Nat Immunol 9(6):658–666 [DOI] [PubMed] [Google Scholar]
- 105.Guy CS, Vignali KM, Temirov J, Bettini ML, Overacre AE, Smeltzer M, Zhang H, Huppa JB, Tsai Y-H, Lobry C, Xie J, Dempsey PJ, Crawford HC, Aifantis I, Davis MM, Vignali DAA (2013) Distinct TCR signaling pathways drive proliferation and cytokine production in T cells. Nat Immunol 14(3):262–270 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 106.James JR (2018) Tuning ITAM multiplicity on T cell receptors can control potency and selectivity to ligand density. Sci Signal 11(531):eaan1088 10.1126/scisignal.aan1088 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 107.Mukhopadhyay H, Cordoba SP, Maini PK, van der Merwe PA, Dushek O (2013) Systems model of T cell receptor proximal signaling reveals emergent ultrasensitivity. PLoS Comput Biol 9(3):e1003004 10.1371/journal.pcbi.1003004 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 108.Salmond RJ, Filby A, Qureshi I, Caserta S, Zamoyska R (2009) T-cell receptor proximal signaling via the Src-family kinases, Lck and Fyn, influences T-cell activation, differentiation, and tolerance. Immunol Rev 228(1):9–22 [DOI] [PubMed] [Google Scholar]
- 109.Stefanova I, Hemmer B, Vergelli M, Martin R, Biddison WE, Germain RN (2003) TCR ligand discrimination is enforced by competing ERK positive and SHP-1 negative feedback pathways. Nat Immunol 4(3):248–254 [DOI] [PubMed] [Google Scholar]
- 110.van Oers NS, Tohlen B, Malissen B, Moomaw CR, Afendis S, Slaughter CA (2000) The 21- and 23-kD forms of TCR zeta are generated by specific ITAM phosphorylations. Nat Immunol 1(4):322–328. 10.1038/79774 [DOI] [PubMed] [Google Scholar]
- 111.Vely F, Nunes JA, Malissen B, Hedgecock CJ (1997) Analysis of immunoreceptor tyrosine-based activation motif (ITAM) binding to ZAP-70 by surface plasmon resonance. Eur J Immunol 27(11):3010–3014. 10.1002/eji.1830271138 [DOI] [PubMed] [Google Scholar]
- 112.Wang H, Kadlecek TA, Au-Yeung BB, Goodfellow HE, Hsu LY, Freedman TS, Weiss A (2010) ZAP-70: an essential kinase in T-cell signaling. Cold Spring Harb Perspect Biol 2:a002279 10.1101/cshperspect.a002279 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 113.Pitcher LA, van Oers NSC (2003) T-cell receptor signal transmission: who gives an ITAM? Trends Immunol 24(10):554–560. 10.1016/j.it.2003.08.003 [DOI] [PubMed] [Google Scholar]
- 114.Courtney AH, Lo WL, Weiss A (2018) TCR signaling: mechanisms of initiation and propagation. Trends Biochem Sci 43(2):108–123. 10.1016/j.tibs.2017.11.008 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 115.Proust R, Bertoglio J, Gesbert F (2012) The adaptor protein SAP directly associates with CD3zeta chain and regulates T cell receptor signaling. PLoS One 7(8):e43200 10.1371/journal.pone.0043200 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 116.Gil D, Schamel WWA, Montoya M, Sanchez-Madrid F, Alarcon B (2002) Recruitment of Nck by CD3 epsilon reveals a ligand-induced conformational change essential for T cell receptor signaling and synapse formation. Cell 109(7):901–912 [DOI] [PubMed] [Google Scholar]
- 117.Kazi JU, Kabir NN, Ronnstrand L (2015) Role of SRC-like adaptor protein (SLAP) in immune and malignant cell signaling. Cell Mol Life Sci 72(13):2535–2544. 10.1007/s00018-015-1882-6 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 118.Sosinowski T, Pandey A, Dixit VM, Weiss A (2000) Src-like adaptor protein (SLAP) is a negative regulator of T cell receptor signaling. J Exp Med 191:463–473 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 119.Myers MD, Dragone LL, Weiss A (2005) Src-like adaptor protein down-regulates T cell receptor (TCR)-CD3 expression by targeting TCRzeta for degradation. J Cell Biol 170(2):285–294. 10.1083/jcb.200501164 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 120.Ksionda O, Saveliev A, Kochl R, Rapley J, Faroudi M, Smith-Garvin JE, Wulfing C, Rittinger K, Carter T, Tybulewicz VLJ (2012) Mechanism and function of Vav1 localisation in TCR signalling. J Cell Sci 125(Pt 22):5302–5314 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 121.Lewis JB, Scangarello FA, Murphy JM, Eidell KP, Sodipo MO, Ophir MJ, Sargeant R, Seminario M-C, Bunnell SC (2018) ADAP is an upstream regulator that precedes SLP-76 at sites of TCR engagement and stabilizes signaling microclusters. J Cell Sci 131(21). 10.1242/jcs.215517 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 122.Yousefi OS, Gunther M, Horner M, Chalupsky J, Wess M, Brandl SM, Smith RW, Fleck C, Kunkel T, Zurbriggen MD, Hofer T, Weber W, Schamel WW (2019) Optogenetic control shows that kinetic proofreading regulates the activity of the T cell receptor. Elife 8 10.7554/eLife.42475 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 123.Stefanova I, Dorfman J, Germain R (2002) Self-recognition promotes the foreign antigen sensitivity of naive T lymphocytes. Nature 420(6914):429–434 [DOI] [PubMed] [Google Scholar]
- 124.Plas DR, Johnson R, Pingel JT, Matthews RJ, Dalton M, Roy G, Chan AC, Thomas ML (1996) Direct regulation of ZAP-70 by SHP-1 in T cell antigen receptor signaling. Science 272(5265):1173–1176 [DOI] [PubMed] [Google Scholar]
- 125.Artyomov MN, Lis M, Devadas S, Davis MM, Chakraborty AK (2010) CD4 and CD8 binding to MHC molecules primarily acts to enhance Lck delivery. Proc Natl Acad Sci U S A 107(39):16916–16921. 10.1073/pnas.1010568107 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 126.Li Q-J, Dinner AR, Qi S, Irvine DJ, Huppa JB, Davis MM, Chakraborty AK (2004) CD4 enhances T cell sensitivity to antigen by coordinating Lck accumulation at the immunological synapse. Nat Immunol 5(8):791–799 [DOI] [PubMed] [Google Scholar]
- 127.Moody AM, Chui D, Reche PA, Priatel JJ, Marth JD, Reinherz EL (2001) Developmentally regulated glycosylation of the CD8alphabeta coreceptor stalk modulates ligand binding. Cell 107(4):501–512 [DOI] [PubMed] [Google Scholar]
- 128.Williams CM, Schonnesen AA, Zhang S-Q, Ma K-Y, He C, Yamamoto T, Eckhardt SG, Klebanoff CA, Jiang N (2017) Normalized synergy predicts that CD8 co-receptor contribution to T cell receptor (TCR) and pMHC binding decreases as TCR affinity increases in human viral-specific T cells. Front Immunol 8:894. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 129.Au-Yeung BB, Levin SE, Zhang C, Hsu L-Y, Cheng DA, Killeen N, Shokat KM, Weiss A (2010) A genetically selective inhibitor demonstrates a function for the kinase Zap70 in regulatory T cells independent of its catalytic activity. Nat Immunol 11(12):1085–1092 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 130.Comrie WA, Burkhardt JK (2016) Action and traction: cytoskeletal control of receptor triggering at the immunological synapse. Front Immunol 7:68. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 131.Steinbuck MP, Arakcheeva K, Winandy S (2018) Novel TCR-mediated mechanisms of notch activation and signaling. J Immunol 200(3):997–1007. 10.4049/jimmunol.1700070 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 132.Wang R, Dillon CP, Shi LZ, Milasta S, Carter R, Finkelstein D, McCormick LL, Fitzgerald P, Chi H, Munger J, Green DR (2011) The transcription factor Myc controls metabolic reprogramming upon T lymphocyte activation. Immunity 35(6):871–882 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 133.Scholz G, Jandus C, Zhang L, Grandclement C, Lopez-Mejia IC, Soneson C, Delorenzi M, Fajas L, Held W, Dormond O, Romero P (2016) Modulation of mTOR signalling triggers the formation of stem cell-like memory T cells. EBioMedicine 4:50–61. 10.1016/j.ebiom.2016.01.019 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 134.Macian F (2005) NFAT proteins: key regulators of T-cell development and function. Nat Rev Immunol 5(6):472–484 [DOI] [PubMed] [Google Scholar]
- 135.Cockerill PN (2016) Receptor signaling directs global recruitment of pre-existing transcription factors to inducible elements. Yale J Biol Med 89(4):591–596 [PMC free article] [PubMed] [Google Scholar]
- 136.Tan MP, Dolton GM, Gerry AB, Brewer JE, Bennett AD, Pumphrey NJ, Jakobsen BK, Sewell AK (2017) Human leucocyte antigen class I-redirected anti-tumour CD4+ T cells require a higher T cell receptor binding affinity for optimal activity than CD8+ T cells. Clin Exp Immunol 187(1):124–137 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 137.de la Roche M, Asano Y, Griffiths GM (2016) Origins of the cytolytic synapse. Nat Rev Immunol 16(7):421–432 [DOI] [PubMed] [Google Scholar]
- 138.Schuijs MJ, Hammad H, Lambrecht BN (2019) Professional and ‘amateur’ antigen-presenting cells in type 2 immunity. Trends Immunol 40(1):22–34 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 139.Lin A, Lore K (2017) Granulocytes: new members of the antigen-presenting cell family. Front Immunol 8:1781. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 140.Rouhani SJ, Eccles JD, Riccardi P, Peske JD, Tewalt EF, Cohen JN, Liblau R, Makinen T, Engelhard VH (2015) Roles of lymphatic endothelial cells expressing peripheral tissue antigens in CD4 T-cell tolerance induction. Nat Commun 6:6771. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 141.Kambayashi T, Laufer TM (2014) Atypical MHC class II-expressing antigen-presenting cells: can anything replace a dendritic cell? Nat Rev Immunol 14(11):719–730 [DOI] [PubMed] [Google Scholar]
- 142.Verboogen DRJ, Dingjan I, Revelo NH, Visser LJ, ter Beest M, van den Bogaart G (2016) The dendritic cell side of the immunological synapse. Biomol Concepts 7(1):17–28 [DOI] [PubMed] [Google Scholar]
- 143.Rodriguez-Fernandez JL, Riol-Blanco L, Delgado-Martin C (2010) What is the function of the dendritic cell side of the immunological synapse? Sci Signal 3(105):re2. [DOI] [PubMed] [Google Scholar]
- 144.Fisher PJ, Bulur PA, Vuk-Pavlovic S, Prendergast FG, Dietz AB (2008) Dendritic cell microvilli: a novel membrane structure associated with the multifocal synapse and T-cell clustering. Blood 112(13):5037–5045 [DOI] [PubMed] [Google Scholar]
- 145.Ronchese F, Hermans IF (2001) Killing of dendritic cells: a life cut short or a purposeful death? J Exp Med 194(5):F23–F26 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 146.Brockman JM, Salaita K (2019) Mechanical proofreading: a general mechanism to enhance the fidelity of information transfer between cells. Front Phys 7 10.3389/fphy.2019.00014 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 147.Legut M, Cole DK, Sewell AK (2015) The promise of gammadelta T cells and the gammadelta T cell receptor for cancer immunotherapy. Cell Mol Immunol 12(6):656–668. 10.1038/cmi.2015.28 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 148.Sharma P, Kranz DM (2016) Recent advances in T-cell engineering for use in immunotherapy. F1000Res 5 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 149.Davenport AJ, Cross RS, Watson KA, Liao Y, Shi W, Prince HM, Beavis PA, Trapani JA, Kershaw MH, Ritchie DS, Darcy PK, Neeson PJ, Jenkins MR (2018) Chimeric antigen receptor T cells form nonclassical and potent immune synapses driving rapid cytotoxicity. Proc Natl Acad Sci U S A 115(9):E2068–E2076 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 150.van der Merwe PA, Dushek O (2011) Mechanisms for T cell receptor triggering. Nat Rev Immunol 11(1):47–55. 10.1038/nri2887 [DOI] [PubMed] [Google Scholar]
- 151.Siller-Farfan JA, Dushek O (2018) Molecular mechanisms of T cell sensitivity to antigen. Immunol Rev 285(1):194–205 [DOI] [PubMed] [Google Scholar]
- 152.Boniface JJ, Rabinowitz JD, Wulfing C, Hampl J, Reich Z, Altman JD, Kantor RM, Beeson C, McConnell HM, Davis MM (1998) Initiation of signal transduction through the T cell receptor requires the multivalent engagement of peptide/MHC ligands [corrected]. Immunity 9(4):459–466 [DOI] [PubMed] [Google Scholar]
- 153.Grossman Z, Paul WE (2015) Dynamic tuning of lymphocytes: physiological basis, mechanisms, and function. Annu Rev Immunol 33:677–713. 10.1146/annurev-immunol-032712-100027 [DOI] [PubMed] [Google Scholar]
- 154.Cho JH, Sprent J (2018) TCR tuning of T cell subsets. Immunol Rev 283(1):129–137. 10.1111/imr.12646 [DOI] [PubMed] [Google Scholar]
- 155.Azzam HS, DeJarnette JB, Huang K, Emmons R, Park CS, Sommers CL, El-Khoury D, Shores EW, Love PE (2001) Fine tuning of TCR signaling by CD5. J Immunol 166(9):5464–5472 [DOI] [PubMed] [Google Scholar]
- 156.Park J-H, Adoro S, Lucas PJ, Sarafova SD, Alag AS, Doan LL, Erman B, Liu X, Ellmeier W, Bosselut R, Feigenbaum L, Singer A (2007) ‘Coreceptor tuning’: cytokine signals transcriptionally tailor CD8 coreceptor expression to the self-specificity of the TCR. Nat Immunol 8(10):1049–1059 [DOI] [PubMed] [Google Scholar]
- 157.Thomas S, Xue SA, Bangham CR, Jakobsen BK, Morris EC, Stauss HJ (2011) Human T cells expressing affinity-matured TCR display accelerated responses but fail to recognize low density of MHC-peptide antigen. Blood 118(2):319–329. 10.1182/blood-2010-12-326736 [DOI] [PubMed] [Google Scholar]
- 158.Junghans V, Santos AM, Lui Y, Davis SJ, Jonsson P (2018) Dimensions and interactions of large T-cell surface proteins. Front Immunol 9:2215. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 159.Jung Y, Riven I, Feigelson SW, Kartvelishvily E, Tohya K, Miyasaka M, Alon R, Haran G (2016) Three-dimensional localization of T-cell receptors in relation to microvilli using a combination of superresolution microscopies. Proc Natl Acad Sci U S A 113(40):E5916–E5924 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 160.Sage PT, Varghese LM, Martinelli R, Sciuto TE, Kamei M, Dvorak AM, Springer TA, Sharpe AH, Carman CV (2012) Antigen recognition is facilitated by invadosome-like protrusions formed by memory/effector T cells. J Immunol 188(8):3686–3699 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 161.Varma R, Campi G, Yokosuka T, Saito T, Dustin ML (2006) T cell receptor-proximal signals are sustained in peripheral microclusters and terminated in the central supramolecular activation cluster. Immunity 25(1):117–127 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 162.Shaw AS, Dustin ML (1997) Making the T cell receptor go the distance: a topological view of T cell activation. Immunity 6(4):361–369 [DOI] [PubMed] [Google Scholar]
- 163.Dustin ML (2014) The immunological synapse. Cancer Immunol Res 2(11): 1023–1033 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 164.Sadreddini S, Baradaran B, Aghebati-Maleki A, Sadreddini S, Shanehbandi D, Fotouhi A, Aghebati-Maleki L (2019) Immune checkpoint blockade opens a new way to cancer immunotherapy. J Cell Physiol 234(6):8541–8549 [DOI] [PubMed] [Google Scholar]
- 165.Parry RV, Chemnitz JM, Frauwirth KA, Lanfranco AR, Braunstein I, Kobayashi SV, Linsley PS, Thompson CB, Riley JL (2005) CTLA-4 and PD-1 receptors inhibit T-cell activation by distinct mechanisms. Mol Cell Biol 25(21):9543–9553. 10.1128/MCB.25.21.9543-9553.2005 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 166.Hui E, Cheung J, Zhu J, Su X, Taylor MJ, Wallweber HA, Sasmal DK, Huang J, Kim JM, Mellman I, Vale RD (2017) T cell costimulatory receptor CD28 is a primary target for PD-1-mediated inhibition. Science 355(6332):1428–1433. 10.1126/science.aaf1292 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 167.Walker LSK (2017) PD-1 and CTLA4: two checkpoints, one pathway? Sci Immunol 2(11). 10.1126/sciimmunol.aan3864 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 168.Walker LSK, Sansom DM (2015) Confusing signals: recent progress in CTLA-4 biology. Trends Immunol 36(2):63–70 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 169.Hivroz C, Chemin K, Tourret M, Bohineust A (2012) Crosstalk between T lymphocytes and dendritic cells. Crit Rev Immunol 32(2):139–155 [DOI] [PubMed] [Google Scholar]
- 170.Watchmaker PB, Urban JA, Berk E, Nakamura Y, Mailliard RB, Watkins SC, van Ham SM, Kalinski P (2008) Memory CD8+ T cells protect dendritic cells from CTL killing. J Immunol 180(6):3857–3865 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 171.Zarour HM (2016) Reversing T-cell dysfunction and exhaustion in cancer. Clin Cancer Res 22(8):1856–1864. 10.1158/1078-0432.CCR-15-1849 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 172.Grywalska E, Pasiarski M, Gozdz S, Rolinski J (2018) Immune-checkpoint inhibitors for combating T-cell dysfunction in cancer. Onco Targets Ther 11:6505–6524. 10.2147/OTT.S150817 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 173.Chung AS, Lee J, Ferrara N (2010) Targeting the tumour vasculature: insights from physiological angiogenesis. Nat Rev Cancer 10(7):505–514. 10.1038/nrc2868 [DOI] [PubMed] [Google Scholar]
- 174.Aptsiauri N, Cabrera T, Mendez R, Garcia-Lora A, Ruiz-Cabello F, Garrido F (2007) Role of altered expression of HLA class I molecules in cancer progression. Adv Exp Med Biol 601:123–131. 10.1007/978-0-387-72005-0_13 [DOI] [PubMed] [Google Scholar]
- 175.Gao D, Jiang L (2018) Exosomes in cancer therapy: a novel experimental strategy. Am J Cancer Res 8(11):2165–2175 [PMC free article] [PubMed] [Google Scholar]
- 176.Andrews MC, Reuben A, Gopalakrishnan V, Wargo JA (2018) Concepts collide: genomic, immune, and microbial influences on the tumor microenvironment and response to cancer therapy. Front Immunol 9:946 10.3389/fimmu.2018.00946 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 177.Pageon SV, Tabarin T, Yamamoto Y, Ma Y, Nicovich PR, Bridgeman JS, Cohnen A, Benzing C, Gao Y, Crowther MD, Tungatt K, Dolton G, Sewell AK, Price DA, Acuto O, Parton RG, Gooding JJ, Rossy J, Rossjohn J, Gaus K (2016) Functional role of T-cell receptor nanoclusters in signal initiation and antigen discrimination. Proc Natl Acad Sci U S A 113(37):E5454–E5463. 10.1073/pnas.1607436113 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 178.Kumar R, Ferez M, Swamy M, Arechaga I, Rejas MT, Valpuesta JM, Schamel WW, Alarcon B, van Santen HM (2011) Increased sensitivity of antigen-experienced T cells through the enrichment of oligomeric T cell receptor complexes. Immunity 35(3):375–387. 10.1016/j.immuni.2011.08.010 [DOI] [PubMed] [Google Scholar]
- 179.Sherman E, Barr V, Manley S, Patterson G, Balagopalan L, Akpan I, Regan CK, Merrill RK, Sommers CL, Lippincott-Schwartz J, Samelson LE (2011) Functional nanoscale organization of signaling molecules downstream of the T cell antigen receptor. Immunity 35(5):705–720. 10.1016/j.immuni.2011.10.004 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 180.Schamel WW, Alarcon B, Hofer T, Minguet S (2017) The allostery model of TCR regulation. J Immunol 198(1):47–52. 10.4049/jimmunol.1601661 [DOI] [PubMed] [Google Scholar]
- 181.Martin-Blanco N, Blanco R, Alda-Catalinas C, Bovolenta ER, Oeste CL, Palmer E, Schamel WW, Lythe G, Molina-Paris C, Castro M, Alarcon B (2018) A window of opportunity for cooperativity in the T cell receptor. Nat Commun 9(1):2618 10.1038/s41467-018-05050-6 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 182.von Essen MR, Kongsbak M, Geisler C (2012) Mechanisms behind functional avidity maturation in T cells. Clin Dev Immunol 2012:163453. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 183.Hayes SM, Love PE (2002) Distinct structure and signaling potential of the gamma delta TCR complex. Immunity 16(6):827–838 [DOI] [PubMed] [Google Scholar]
- 184.Hayes SM, Love PE (2006) Stoichiometry of the murine gammadelta T cell receptor. J Exp Med 203(1):47–52. 10.1084/jem.20051886 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 185.Dave VP, Cao Z, Browne C et al. (1997) CD3 delta deficiency arrests development of the alpha beta but not the gamma delta T cell lineage. EMBO J 16(6):1360–1370 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 186.Siegers GM, Swamy M, Fernandez-Malave E, Minguet S, Rathmann S, Guardo AC, Perez-Flores V, Regueiro JR, Alarcon B, Fisch P, Schamel WW (2007) Different composition of the human and the mouse gammadelta T cell receptor explains different phenotypes of CD3gamma and CD3delta immunodeficiencies. J Exp Med 204(11):2537–2544. 10.1084/jem.20070782 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 187.Cheng HY, Wu R, Gebre AK, Hanna RN, Smith DJ, Parks JS, Ley K, Hedrick CC (2013) Increased cholesterol content in gammadelta (gammadelta) T lymphocytes differentially regulates their activation. PLoS One 8(5):e63746 10.1371/journal.pone.0063746 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 188.Haks MC, Lefebvre JM, Lauritsen JPH, Carleton M, Rhodes M, Miyazaki T, Kappes DJ, Wiest DL (2005) Attenuation of gammadeltaTCR signaling efficiently diverts thymocytes to the alphabeta lineage. Immunity 22(5):595–606 [DOI] [PubMed] [Google Scholar]
- 189.Nicolas L, Monneret G, Debard AL, Blesius A, Gutowski MC, Salles G, Bienvenu J (2001) Human gammadelta T cells express a higher TCR/CD3 complex density than alphabeta T cells. Clin Immunol 98(3):358–363 [DOI] [PubMed] [Google Scholar]
- 190.Hayes SM, Li L, Love PE (2005) TCR signal strength influences alphabeta/gammadelta lineage fate. Immunity 22(5):583–593 [DOI] [PubMed] [Google Scholar]
- 191.Dopfer EP, Hartl FA, Oberg HH, Siegers GM, Yousefi OS, Kock S, Fiala GJ, Garcillan B, Sandstrom A, Alarcon B, Regueiro JR, Kabelitz D, Adams EJ, Minguet S, Wesch D, Fisch P, Schamel WWA (2014) The CD3 conformational change in the gammadelta T cell receptor is not triggered by antigens but can be enforced to enhance tumor killing. Cell Rep 7(5):1704–1715. 10.1016/j.celrep.2014.04.049 [DOI] [PubMed] [Google Scholar]
- 192.Muro R, Takayanagi H, Nitta T (2019) T cell receptor signaling for gammadeltaT cell development. Inflamm Regen 39:6 10.1186/s41232-019-0095-z [DOI] [PMC free article] [PubMed] [Google Scholar]
- 193.Laird RM, Laky K, Hayes SM (2010) Unexpected role for the B cell-specific Src family kinase B lymphoid kinase in the development of IL-17-producing gammadelta T cells. J Immunol 185(11):6518–6527. 10.4049/jimmunol.1002766 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 194.Getts D, Hofmeister R, Quintas-Cardama A (2019) Synthetic T cell receptor-based lymphocytes for cancer therapy. Adv Drug Deliv Rev. 10.1016/j.addr.2019.04.002 [DOI] [PubMed] [Google Scholar]
- 195.Thomas S, Stauss HJ, Morris EC (2010) Molecular immunology lessons from therapeutic T-cell receptor gene transfer. Immunology 129(2):170–177 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 196.van Loenen MM, de Boer R, Amir AL, Hagedoorn RS, Volbeda GL, Willemze R, van Rood JJ, Falkenburg JHF, Heemskerk MHM (2010) Mixed T cell receptor dimers harbor potentially harmful neoreactivity. Proc Natl Acad Sci U S A 107(24):10972–10977 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 197.Voss R-H, Willemsen RA, Kuball J, Grabowski M, Engel R, Intan RS, Guillaume P, Romero P, Huber C, Theobald M (2008) Molecular design of the Calphabeta interface favors specific pairing of introduced TCRalphabeta in human T cells. J Immunol 180(1):391–401 [DOI] [PubMed] [Google Scholar]
- 198.Sommermeyer D, Uckert W (2010) Minimal amino acid exchange in human TCR constant regions fosters improved function of TCR gene-modified T cells. J Immunol 184(11):6223–6231 [DOI] [PubMed] [Google Scholar]
- 199.Cohen CJ, Zhao Y, Zheng Z, Rosenberg SA, Morgan RA (2006) Enhanced antitumor activity of murine-human hybrid T-cell receptor (TCR) in human lymphocytes is associated with improved pairing and TCR/CD3 stability. Cancer Res 66(17):8878–8886 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 200.Rosati S, Parkhurst MR, Hong Y et al. (2014) A novel murine T-cell receptor targeting NY-ESO-1. J Immunother 37:135–146 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 201.Legut M, Dolton G, Mian AA, Ottmann OG, Sewell AK (2018) CRISPR-mediated TCR replacement generates superior anticancer transgenic T cells. Blood 131(3): 311–322. 10.1182/blood-2017-05-787598 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 202.Ahmadi M, King JW, Xue S-A, Voisine C, Holler A, Wright GP, Waxman J, Morris E, Stauss HJ (2011) CD3 limits the efficacy of TCR gene therapy in vivo. Blood 118(13):3528–3537 [DOI] [PubMed] [Google Scholar]
- 203.Liddy N, Bossi G, Adams KJ, Lissina A, Mahon TM, Hassan NJ, Gavarret J, Bianchi FC, Pumphrey NJ, Ladell K, Gostick E, Sewell AK, Lissin NM, Harwood NE, Molloy PE, Li Y, Cameron BJ, Sami M, Baston EE, Todorov PT, Paston SJ, Dennis RE, Harper JV, Dunn SM, Ashfield R, Johnson A, McGrath Y, Plesa G, June CH, Kalos M, Price DA, Vuidepot A, Williams DD, Sutton DH, Jakobsen BK (2012) Monoclonal TCR-redirected tumor cell killing. Nat Med 18(6):980–987 [DOI] [PubMed] [Google Scholar]
- 204.Rapoport AP, Stadtmauer EA, Binder-Scholl GK, Goloubeva O, Vogl DT, Lacey SF, Badros AZ, Garfall A, Weiss B, Finklestein J, Kulikovskaya I, Sinha SK, Kronsberg S, Gupta M, Bond S, Melchiori L, Brewer JE, Bennett AD, Gerry AB, Pumphrey NJ, Williams D, Tayton-Martin HK, Ribeiro L, Holdich T, Yanovich S, Hardy N, Yared J, Kerr N, Philip S, Westphal S, Siegel DL, Levine BL, Jakobsen BK, Kalos M, June CH (2015) NY-ESO-1-specific TCR-engineered T cells mediate sustained antigen-specific antitumor effects in myeloma. Nat Med 21(8):914–921. 10.1038/nm.3910 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 205.Kang S, Li Y, Bao Y, Li Y (2019) High-affinity T cell receptors redirect cytokine-activated T cells (CAT) to kill cancer cells. Front Med 13(1):69–82. 10.1007/s11684-018-0677-1 [DOI] [PubMed] [Google Scholar]
- 206.Matsuda T, Leisegang M, Park J-H, Ren L, Kato T, Ikeda Y, Harada M, Kiyotani K, Lengyel E, Fleming GF, Nakamura Y (2018) Induction of neoantigen-specific cytotoxic T cells and construction of T-cell receptor-engineered T cells for ovarian cancer. Clin Cancer Res 24(21):5357–5367 [DOI] [PubMed] [Google Scholar]
- 207.Kato T, Matsuda T, Ikeda Y, Park J-H, Leisegang M, Yoshimura S, Hikichi T, Harada M, Zewde M, Sato S, Hasegawa K, Kiyotani K, Nakamura Y (2018) Effective screening of T cells recognizing neoantigens and construction of T-cell receptor-engineered T cells. Oncotarget 9(13):11009–11019 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 208.Gross G, Waks T, Eshhar Z (1989) Expression of immunoglobulin-T-cell receptor chimeric molecules as functional receptors with antibody-type specificity. Proc Natl Acad Sci U S A 86(24):10024–10028 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 209.Duan H, Huang H, Jing G (2019) An antibody Fab fragment-based chimeric antigen receptor could efficiently eliminate human thyroid cancer cells. J Cancer 10(8):1890–1895. 10.7150/jca.30163 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 210.Gross G, Eshhar Z (2016) Therapeutic potential of T cell chimeric antigen receptors (CARs) in cancer treatment: counteracting off-tumor toxicities for safe CAR T cell therapy. Annu Rev Pharmacol Toxicol 56:59–83 [DOI] [PubMed] [Google Scholar]
- 211.Ramos CA, Dotti G (2011) Chimeric antigen receptor (CAR)-engineered lymphocytes for cancer therapy. Expert Opin Biol Ther 11(7):855–873. 10.1517/14712598.2011.573476 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 212.Rodgers DT, Mazagova M, Hampton EN, Cao Y, Ramadoss NS, Hardy IR, Schulman A, Du J, Wang F, Singer O, Ma J, Nunez V, Shen J, Woods AK, Wright TM, Schultz PG, Kim CH, Young TS (2016) Switch-mediated activation and retargeting of CAR-T cells for B-cell malignancies. Proc Natl Acad Sci U S A 113(4):E459–E468. 10.1073/pnas.1524155113 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 213.Romeo C, Marine A, Seed B (1992) Sequence requirements for induction of cytolysis by the T cell antigen/Fc receptor zeta chain. Cell 68:889–897 [DOI] [PubMed] [Google Scholar]
- 214.Lim WA, June CH (2017) The principles of engineering immune cells to treat cancer. Cell 168(4):724–740. 10.1016/j.cell.2017.01.016 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 215.Dodson LF, Boomer JS, Deppong CM, Shah DD, Sim J, Bricker TL, Russell JH, Green JM (2009) Targeted knock-in mice expressing mutations of CD28 reveal an essential pathway for costimulation. Mol Cell Biol 29(13):3710–3721. 10.1128/MCB.01869-08 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 216.Boomer JS, Green JM (2010) An enigmatic tail of CD28 signaling. Cold Spring Harb Perspect Biol 2(8):a002436 10.1101/cshperspect.a002436 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 217.Wu LX, La Rose J, Chen L, Neale C, Mak T, Okkenhaug K, Wange R, Rottapel R (2005) CD28 regulates the translation of Bcl-xL via the phosphatidylinositol 3-kinase/mammalian target of rapamycin pathway. J Immunol 174(1):180–194. 10.4049/jimmunol.174.1.180 [DOI] [PubMed] [Google Scholar]
- 218.Zapata JM, Perez-Chacon G, Carr-Baena P, Martinez-Forero I, Azpilikueta A, Otano I, Melero I (2018) CD137 (4–1BB) signalosome: complexity is a matter of TRAFs. Front Immunol 9:2618 10.3389/fimmu.2018.02618 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 219.Nam KO, Kang H, Shin SM, Cho KH, Kwon B, Kwon BS, Kim SJ, Lee HW (2005) Cross-linking of 4–1BB activates TCR-signaling pathways in CD8+ T lymphocytes. J Immunol 174(4):1898–1905. 10.4049/jimmunol.174.4.1898 [DOI] [PubMed] [Google Scholar]
- 220.Abate-Daga D, Davila ML (2016) CAR models: next-generation CAR modifications for enhanced T-cell function. Mol Ther Oncolytics 3:16014 10.1038/mto.2016.14 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 221.Guedan S, Calderon H, Posey AD Jr, Maus MV (2019) Engineering and design of chimeric antigen receptors. Mol Ther Methods Clin Dev 12:145–156. 10.1016/j.omtm.2018.12.009 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 222.Weinkove R, George P, Dasyam N, McLellan AD (2019) Selecting costimulatory domains for chimeric antigen receptors: functional and clinical considerations. Clin Transl Immunol 8(5):e1049 10.1002/cti2.1049 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 223.Sadelain M, Brentjens R, Riviere I (2013) The basic principles of chimeric antigen receptor design. Cancer Discov 3(4):388–398 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 224.Shirasu N, Yamada H, Shibaguchi H, Kuroki M, Kuroki M (2015) Corrigendum to “Molecular characterization of a fully human chimeric T-cell antigen receptor for tumor-associated antigen EpCAM”. Biomed Res Int 2015:292436. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 225.Kalaitsidou M, Kueberuwa G, Schutt A, Gilham DE (2015) CAR T-cell therapy: toxicity and the relevance of preclinical models. Immunotherapy 7(5):487–497 [DOI] [PubMed] [Google Scholar]
- 226.Hermanson DL, Kaufman DS (2015) Utilizing chimeric antigen receptors to direct natural killer cell activity. Front Immunol 6:195 10.3389/fimmu.2015.00195 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 227.Wang D, Aguilar B, Starr R, Alizadeh D, Brito A, Sarkissian A, Ostberg JR, Forman SJ, Brown CE (2018) Glioblastoma-targeted CD4+ CAR T cells mediate superior antitumor activity. JCI Insight 3(10). 10.1172/jci.insight.99048 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 228.Hong LK, Chen Y, Smith CC, Montgomery SA, Vincent BG, Dotti G, Savoldo B (2018) CD30-redirected chimeric antigen receptor T cells target CD30(+) and CD30(−) embryonal carcinoma via antigen-dependent and Fas/FasL interactions. Cancer Immunol Res 6(10):1274–1287. 10.1158/2326-6066.CIR-18-0065 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 229.Rabinovich PM, Komarovskaya ME, Wrzesinski SH, Alderman JL, Budak-Alpdogan T, Karpikov A, Guo H, Flavell RA, Cheung NK, Weissman SM, Bahceci E (2009) Chimeric receptor mRNA transfection as a tool to generate antineoplastic lymphocytes. Hum Gene Ther 20(1):51–61. 10.1089/hum.2008.068 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 230.Du Y, Wei Y (2018) Therapeutic potential of natural killer cells in gastric cancer. Front Immunol 9:3095 10.3389/fimmu.2018.03095 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 231.Bollino D, Webb TJ (2017) Chimeric antigen receptor-engineered natural killer and natural killer T cells for cancer immunotherapy. Transl Res 187:32–43. 10.1016/j.trsl.2017.06.003 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 232.Fisher J, Anderson J (2018) Engineering approaches in human gamma delta T cells for cancer immunotherapy. Front Immunol 9:1409 10.3389/fimmu.2018.01409 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 233.Capsomidis A, Benthall G, Van Acker HH, Fisher J, Kramer AM, Abeln Z, Majani Y, Gileadi T, Wallace R, Gustafsson K, Flutter B, Anderson J (2018) Chimeric antigen receptor-engineered human gamma delta T cells: enhanced cytotoxicity with retention of cross presentation. Mol Ther 26(2):354–365. 10.1016/j.ymthe.2017.12.001 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 234.Kriegsmann K, Kriegsmann M, von Bergwelt-Baildon M, Cremer M, Witzens-Harig M (2018) NKT cells - new players in CAR cell immunotherapy? Eur J Haematol 101(6):750–757. 10.1111/ejh.13170 [DOI] [PubMed] [Google Scholar]
- 235.Simon B, Wiesinger M, Marz J, Wistuba-Hamprecht K, Weide B, Schuler-Thurner B, Schuler G, Dorrie J, Uslu U (2018) The generation of CAR-transfected natural killer T cells for the immunotherapy of melanoma. Int J Mol Sci 19(8). 10.3390/ijms19082365 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 236.Roberts MR, Cooke KS, Tran AC, Smith KA, Lin WY, Wang M, Dull TJ, Farson D, Zsebo KM, Finer MH (1998) Antigen-specific cytolysis by neutrophils and NK cells expressing chimeric immune receptors bearing zeta or gamma signaling domains. J Immunol 161(1):375–384 [PubMed] [Google Scholar]
- 237.Liu X, Jiang S, Fang C, Yang S, Olalere D, Pequignot EC, Cogdill AP, Li N, Ramones M, Granda B, Zhou L, Loew A, Young RM, June CH, Zhao Y (2015) Affinity-tuned ErbB2 or EGFR chimeric antigen receptor T cells exhibit an increased therapeutic index against tumors in mice. Cancer Res 75(17):3596–3607. 10.1158/0008-5472.CAN-15-0159 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 238.Park S, Shevlin E, Vedvyas Y, Zaman M, Park S, Hsu YS, Min IM, Jin MM (2017) Micromolar affinity CAR T cells to ICAM-1 achieves rapid tumor elimination while avoiding systemic toxicity. Sci Rep 7(1):14366 10.1038/s41598-017-14749-3 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 239.Drent E, Themeli M, Poels R, de Jong-Korlaar R, Yuan H, de Bruijn J, Martens ACM, Zweegman S, van de Donk N, Groen RWJ, Lokhorst HM, Mutis T (2017) A rational strategy for reducing on-target off-tumor effects of CD38-chimeric antigen receptors by affinity optimization. Mol Ther 25(8):1946–1958. 10.1016/j.ymthe.2017.04.024 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 240.Ma JS, Kim JY, Kazane SA, Choi SH, Yun HY, Kim MS, Rodgers DT, Pugh HM, Singer O, Sun SB, Fonslow BR, Kochenderfer JN, Wright TM, Schultz PG, Young TS, Kim CH, Cao Y (2016) Versatile strategy for controlling the specificity and activity of engineered T cells. Proc Natl Acad Sci U S A 113(4):E450–E458. 10.1073/pnas.1524193113 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 241.Ying Z, Huang XF, Xiang X, Liu Y, Kang X, Song Y, Guo X, Liu H, Ding N, Zhang T, Duan P, Lin Y, Zheng W, Wang X, Lin N, Tu M, Xie Y, Zhang C, Liu W, Deng L, Gao S, Ping L, Wang X, Zhou N, Zhang J, Wang Y, Lin S, Mamuti M, Yu X, Fang L, Wang S, Song H, Wang G, Jones L, Zhu J, Chen SY (2019) A safe and potent anti-CD19 CAR T cell therapy. Nat Med. 10.1038/s41591-019-0421-7 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 242.Kumaresan PR, Manuri PR, Albert ND, Maiti S, Singh H, Mi T, Roszik J, Rabinovich B, Olivares S, Krishnamurthy J, Zhang L, Najjar AM, Huls MH, Lee DA, Champlin RE, Kontoyiannis DP, Cooper LJN (2014) Bioengineering T cells to target carbohydrate to treat opportunistic fungal infection. Proc Natl Acad Sci U S A 111(29):10660–10665 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 243.Townsend MH, Shrestha G, Robison RA, O’Neill KL (2018) The expansion of targetable biomarkers for CAR T cell therapy. J Exp Clin Cancer Res 37(1):163 10.1186/s13046-018-0817-0 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 244.Shaffer DR, Savoldo B, Yi Z, Chow KKH, Kakarla S, Spencer DM, Dotti G, Wu M-F, Liu H, Kenney S, Gottschalk S (2011) T cells redirected against CD70 for the immunotherapy of CD70-positive malignancies. Blood 117(16):4304–4314 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 245.Urbanska K, Lanitis E, Poussin M, Lynn RC, Gavin BP, Kelderman S, Yu J, Scholler N, Powell DJ Jr (2012) A universal strategy for adoptive immunotherapy of cancer through use of a novel T-cell antigen receptor. Cancer Res 72(7):1844–1852 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 246.Liu K, Liu X, Peng Z, Sun H, Zhang M, Zhang J, Liu S, Hao L, Lu G, Zheng K, Gong X, Wu D, Wang F, Shen L (2015) Retargeted human avidin-CAR T cells for adoptive immunotherapy of EGFRvIII expressing gliomas and their evaluation via optical imaging. Oncotarget 6(27):23735–23747 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 247.Bridgeman JS, Ladell K, Sheard VE, Miners K, Hawkins RE, Price DA, Gilham DE (2014) CD3zeta-based chimeric antigen receptors mediate T cell activation via cis- and trans-signalling mechanisms: implications for optimization of receptor structure for adoptive cell therapy. Clin Exp Immunol 175(2):258–267. 10.1111/cei.12216 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 248.Watanabe K, Terakura S, Martens AC, van Meerten T, Uchiyama S, Imai M, Sakemura R, Goto T, Hanajiri R, Imahashi N, Shimada K, Tomita A, Kiyoi H, Nishida T, Naoe T, Murata M (2015) Target antigen density governs the efficacy of anti-CD20-CD28-CD3 zeta chimeric antigen receptor-modified effector CD8+ T cells. J Immunol 194(3):911–920. 10.4049/jimmunol.1402346 [DOI] [PubMed] [Google Scholar]
- 249.Stone JD, Aggen DH, Schietinger A, Schreiber H, Kranz DM (2012) A sensitivity scale for targeting T cells with chimeric antigen receptors (CARs) and bispecific T-cell Engagers (BiTEs). Oncoimmunology 1(6):863–873. 10.4161/onci.20592 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 250.Long AH, Haso WM, Shern JF, Wanhainen KM, Murgai M, Ingaramo M, Smith JP, Walker AJ, Kohler ME, Venkateshwara VR, Kaplan RN, Patterson GH, Fry TJ, Orentas RJ, Mackall CL (2015) 4–1BB costimulation ameliorates T cell exhaustion induced by tonic signaling of chimeric antigen receptors. Nat Med 21(6):581–590. 10.1038/nm.3838 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 251.Gomes-Silva D, Mukherjee M, Srinivasan M, Krenciute G, Dakhova O, Zheng Y, Cabral JMS, Rooney CM, Orange JS, Brenner MK, Mamonkin M (2017) Tonic 4–1BB costimulation in chimeric antigen receptors impedes T cell survival and is vector-dependent. Cell Rep 21(1):17–26 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 252.Ajina A, Maher J (2018) Strategies to address chimeric antigen receptor tonic signaling. Mol Cancer Ther 17(9):1795–1815 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 253.Watanabe N, Bajgain P, Sukumaran S, Ansari S, Heslop HE, Rooney CM, Brenner MK, Leen AM, Vera JF (2016) Fine-tuning the CAR spacer improves T-cell potency. Oncoimmunology 5(12):e1253656 10.1080/2162402X.2016.1253656 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 254.Feucht J, Sun J, Eyquem J, Ho YJ, Zhao Z, Leibold J, Dobrin A, Cabriolu A, Hamieh M, Sadelain M (2019) Calibration of CAR activation potential directs alternative T cell fates and therapeutic potency. Nat Med 25(1):82–88. 10.1038/s41591-018-0290-5 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 255.Tschumi BO, Dumauthioz N, Marti B, Zhang L, Schneider P, Mach JP, Romero P, Donda A (2018) CART cells are prone to Fas- and DR5-mediated cell death. J Immunother Cancer 6(1):71 10.1186/s40425-018-0385-z [DOI] [PMC free article] [PubMed] [Google Scholar]
- 256.Grupp SA, Kalos M, Barrett D, Aplenc R, Porter DL, Rheingold SR, Teachey DT, Chew A, Hauck B, Wright JF, Milone MC, Levine BL, June CH (2013) Chimeric antigen receptor-modified T cells for acute lymphoid leukemia. N Engl J Med 368(16):1509–1518. 10.1056/NEJMoa1215134 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 257.Porter DL, Levine BL, Kalos M, Bagg A, June CH (2011) Chimeric antigen receptor-modified T cells in chronic lymphoid leukemia. N Engl J Med 365(8):725–733 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 258.Teachey DT, Lacey SF, Shaw PA, Melenhorst JJ, Maude SL, Frey N, Pequignot E, Gonzalez VE, Chen F, Finklestein J, Barrett DM, Weiss SL, Fitzgerald JC, Berg RA, Aplenc R, Callahan C, Rheingold SR, Zheng Z, Rose-John S, White JC, Nazimuddin F, Wertheim G, Levine BL, June CH, Porter DL, Grupp SA (2016) Identification of predictive biomarkers for cytokine release syndrome after chimeric antigen receptor T-cell therapy for acute lymphoblastic leukemia. Cancer Discov 6(6):664–679 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 259.Uttenthal BJ, Chua I, Morris EC, Stauss HJ (2012) Challenges in T cell receptor gene therapy. J Gene Med 14(6):386–399 [DOI] [PubMed] [Google Scholar]
- 260.Lee DW, Kochenderfer JN, Stetler-Stevenson M, Cui YK, Delbrook C, Feldman SA, Fry TJ, Orentas R, Sabatino M, Shah NN, Steinberg SM, Stroncek D, Tschernia N, Yuan C, Zhang H, Zhang L, Rosenberg SA, Wayne AS, Mackall CL (2015) T cells expressing CD19 chimeric antigen receptors for acute lymphoblastic leukaemia in children and young adults: a phase 1 dose-escalation trial. Lancet 385(9967):517–528 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 261.Evans AG, Rothberg PG, Burack WR, Huntington SF, Porter DL, Friedberg JW, Liesveld JL (2015) Evolution to plasmablastic lymphoma evades CD19-directed chimeric antigen receptor T cells. Br J Haematol 171(2):205–209 [DOI] [PubMed] [Google Scholar]
- 262.Maude SL, Frey N, Shaw PA, Aplenc R, Barrett DM, Bunin NJ, Chew A, Gonzalez VE, Zheng Z, Lacey SF, Mahnke YD, Melenhorst JJ, Rheingold SR, Shen A, Teachey DT, Levine BL, June CH, Porter DL, Grupp SA (2014) Chimeric antigen receptor T cells for sustained remissions in leukemia. N Engl J Med 371(16):1507–1517 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 263.Cho JH, Collins JJ, Wong WW (2018) Universal chimeric antigen receptors for multiplexed and logical control of T cell responses. Cell 173(6):1426–1438.e1411. 10.1016/j.cell.2018.03.038 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 264.Walseng E, Koksal H, Sektioglu IM, Fane A, Skorstad G, Kvalheim G, Gaudernack G, Inderberg EM, Walchli S (2017) A TCR-based chimeric antigen receptor. Sci Rep 7(1):10713 10.1038/s41598-017-11126-y [DOI] [PMC free article] [PubMed] [Google Scholar]
- 265.Helsen CW, Hammill JA, Lau VWC, Mwawasi KA, Afsahi A, Bezverbnaya K, Newhook L, Hayes DL, Aarts C, Bojovic B, Denisova GF, Kwiecien JM, Brain I, Derocher H, Milne K, Nelson BH, Bramson JL (2018) The chimeric TAC receptor co-opts the T cell receptor yielding robust anti-tumor activity without toxicity. Nat Commun 9(1):3049. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 266.Baeuerle PA, Ding J, Patel E, Thorausch N, Horton H, Gierut J, Scarfo I, Choudhary R, Kiner O, Krishnamurthy J, Le B, Morath A, Baldeviano GC, Quinn J, Tavares P, Wei Q, Weiler S, Maus MV, Getts D, Schamel WW, Hofmeister R (2019) Synthetic TRuC receptors engaging the complete T cell receptor for potent anti-tumor response. Nat Commun 10(1):2087 10.1038/s41467-019-10097-0 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 267.DeFord-Watts LM, Young JA, Pitcher LA, van Oers NSC (2007) The membrane-proximal portion of CD3 epsilon associates with the serine/threonine kinase GRK2. J Biol Chem 282(22):16126–16134 [DOI] [PubMed] [Google Scholar]
- 268.Yamazaki T, Hamano Y, Tashiro H, Itoh K, Nakano H, Miyatake S, Saito T (1999) CAST, a novel CD3epsilon-binding protein transducing activation signal for interleukin-2 production in T cells. J Biol Chem 274(26):18173–18180 [DOI] [PubMed] [Google Scholar]
- 269.Deford-Watts LM, Tassin TC, Becker AM, Medeiros JJ, Albanesi JP, Love PE, Wulfing C, van Oers NSC (2009) The cytoplasmic tail of the T cell receptor CD3 epsilon subunit contains a phospholipid-binding motif that regulates T cell functions. J Immunol 183(2):1055–1064 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 270.Kagoya Y, Tanaka S, Guo T, Anczurowski M, Wang CH, Saso K, Butler MO, Minden MD, Hirano N (2018) A novel chimeric antigen receptor containing a JAK-STAT signaling domain mediates superior antitumor effects. Nat Med 24(3):352–359. 10.1038/nm.4478 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 271.Roybal KT, Rupp LJ, Morsut L, Walker WJ, McNally KA, Park JS, Lim WA (2016) Precision tumor recognition by T cells with combinatorial antigen-sensing circuits. Cell 164(4):770–779 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 272.Shah NN, Maatman T, Hari P, Johnson B (2019) Multi targeted CAR-T cell therapies for B-cell malignancies. Front Oncol 9:146 10.3389/fonc.2019.00146 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 273.D’Aloia MM, Zizzari IG, Sacchetti B, Pierelli L, Alimandi M (2018) CAR-T cells: the long and winding road to solid tumors. Cell Death Dis 9(3):282 10.1038/s41419-018-0278-6 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 274.Bagashev A, Sotillo E, Tang C-HA, Black KL, Perazzelli J, Seeholzer SH, Argon Y, Barrett DM, Grupp SA, Hu C-CA, Thomas-Tikhonenko A (2018) CD19 alterations emerging after CD19-directed immunotherapy cause retention of the misfolded protein in the endoplasmic reticulum. Mol Cell Biol 38(21):e00383–e00318 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 275.Schneider D, Xiong Y, Wu D, Nölle V, Schmitz S, Haso W, Kaiser A, Dropulic B, Orentas RJ (2017) A tandem CD19/CD20 CAR lentiviral vector drives on-target and off-target antigen modulation in leukemia cell lines. J Immunother Cancer 5:42. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 276.Hegde M, Mukherjee M, Grada Z, Pignata A, Landi D, Navai SA, Wakefield A, Fousek K, Bielamowicz K, Chow KK, Brawley VS, Byrd TT, Krebs S, Gottschalk S, Wels WS, Baker ML, Dotti G, Mamonkin M, Brenner MK, Orange JS, Ahmed N (2016) Tandem CAR T cells targeting HER2 and IL13Ralpha2 mitigate tumor antigen escape. J Clin Invest 126(8):3036–3052. 10.1172/JCI83416 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 277.Kloss CC, Condomines M, Cartellieri M, Bachmann M, Sadelain M (2013) Combinatorial antigen recognition with balanced signaling promotes selective tumor eradication by engineered T cells. Nat Biotechnol 31(1):71–75 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 278.Fedorov VD, Themeli M, Sadelain M (2013) PD-1- and CTLA-4-based inhibitory chimeric antigen receptors (iCARs) divert off-target immunotherapy responses. Sci Transl Med 5(215):215ra172 10.1126/scitranslmed.3006597 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 279.Liu X, Ranganathan R, Jiang S, Fang C, Sun J, Kim S, Newick K, Lo A, June CH, Zhao Y, Moon EK (2016) A chimeric switch-receptor targeting PD1 augments the efficacy of second-generation CAR T cells in advanced solid tumors. Cancer Res 76(6):1578–1590 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 280.Ankri C, Shamalov K, Horovitz-Fried M, Mauer S, Cohen CJ (2013) Human T cells engineered to express a programmed death 1/28 costimulatory retargeting molecule display enhanced antitumor activity. J Immunol 191(8):4121–4129 [DOI] [PubMed] [Google Scholar]
- 281.Kobold S, Grassmann S, Chaloupka M, Lampert C, Wenk S, Kraus F, Rapp M, Duwell P, Zeng Y, Schmollinger JC, Schnurr M, Endres S, RothenfuSser S (2015) Impact of a new fusion receptor on PD-1-mediated immunosuppression in adoptive T cell therapy. J Natl Cancer Inst 107(8):djv146. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 282.Prosser ME, Brown CE, Shami AF, Forman SJ, Jensen MC (2012) Tumor PD-L1 co-stimulates primary human CD8(+) cytotoxic T cells modified to express a PD1:CD28 chimeric receptor. Mol Immunol 51(3–4):263–272. 10.1016/j.molimm.2012.03.023 [DOI] [PubMed] [Google Scholar]
- 283.Chmielewski M, Abken H (2015) TRUCKs: the fourth generation of CARs. Expert Opin Biol Ther 15(8):1145–1154. 10.1517/14712598.2015.1046430 [DOI] [PubMed] [Google Scholar]
- 284.Chmielewski M, Kopecky C, Hombach AA, Abken H (2011) IL-12 release by engineered T cells expressing chimeric antigen receptors can effectively Muster an antigen-independent macrophage response on tumor cells that have shut down tumor antigen expression. Cancer Res 71(17):5697–5706 [DOI] [PubMed] [Google Scholar]
- 285.Yeku OO, Brentjens RJ (2016) Armored CAR T-cells: utilizing cytokines and proinflammatory ligands to enhance CAR T-cell anti-tumour efficacy. Biochem Soc Trans 44(2):412–418 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 286.Avanzi MP, Yeku O, Li X, Wijewarnasuriya DP, van Leeuwen DG, Cheung K, Park H, Purdon TJ, Daniyan AF, Spitzer MH, Brentjens RJ (2018) Engineered tumor-targeted T cells mediate enhanced anti-tumor efficacy both directly and through activation of the endogenous immune system. Cell Rep 23(7):2130–2141 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 287.Adachi K, Kano Y, Nagai T, Okuyama N, Sakoda Y, Tamada K (2018) IL-7 and CCL19 expression in CAR-T cells improves immune cell infiltration and CAR-T cell survival in the tumor. Nat Biotechnol 36(4):346–351. 10.1038/nbt.4086 [DOI] [PubMed] [Google Scholar]
- 288.Shum T, Kruse RL, Rooney CM (2018) Strategies for enhancing adoptive T-cell immunotherapy against solid tumors using engineered cytokine signaling and other modalities. Expert Opin Biol Ther 18(6):653–664 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 289.Curran KJ, Seinstra BA, Nikhamin Y, Yeh R, Usachenko Y, van Leeuwen DG, Purdon T, Pegram HJ, Brentjens RJ (2015) Enhancing antitumor efficacy of chimeric antigen receptor T cells through constitutive CD40L expression. Mol Ther 23(4):769–778 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 290.Rafiq S, Yeku OO, Jackson HJ, Purdon TJ, van Leeuwen DG, Drakes DJ, Song M, Miele MM, Li Z, Wang P, Yan S, Xiang J, Ma X, Seshan VE, Hendrickson RC, Liu C, Brentjens RJ (2018) Targeted delivery of a PD-1-blocking scFv by CAR-T cells enhances anti-tumor efficacy in vivo. Nat Biotechnol 36(9):847–856 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 291.Di Stasi A, Tey S-K, Dotti G, Fujita Y, Kennedy-Nasser A, Martinez C, Straathof K, Liu E, Durett AG, Grilley B, Liu H, Cruz CR, Savoldo B, Gee AP, Schindler J, Krance RA, Heslop HE, Spencer DM, Rooney CM, Brenner MK (2011) Inducible apoptosis as a safety switch for adoptive cell therapy. N Engl J Med 365(18):1673–1683 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 292.Roybal KT, Williams JZ, Morsut L, Rupp LJ, Kolinko I, Choe JH, Walker WJ, McNally KA, Lim WA (2016) Engineering T cells with customized therapeutic response programs using synthetic notch receptors. Cell 167(2):419–432.e416 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 293.Kamiya T, Wong D, Png YT, Campana D (2018) A novel method to generate T-cell receptor-deficient chimeric antigen receptor T cells. Blood Adv 2(5):517–528 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 294.Legrand F, Driss V, Woerly G, Loiseau S, Hermann E, Fournie JJ, Heliot L, Mattot V, Soncin F, Gougeon ML, Dombrowicz D, Capron M (2009) A functional gammadeltaTCR/CD3 complex distinct from gammadeltaT cells is expressed by human eosinophils. PLoS One 4(6):e5926 10.1371/journal.pone.0005926 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 295.Beham AW, Puellmann K, Laird R, Fuchs T, Streich R, Breysach C, Raddatz D, Oniga S, Peccerella T, Findeisen P, Kzhyshkowska J, Gratchev A, Schweyer S, Saunders B, Wessels JT, Mobius W, Keane J, Becker H, Ganser A, Neumaier M, Kaminski WE (2011) A TNF-regulated recombinatorial macrophage immune receptor implicated in granuloma formation in tuberculosis. PLoS Pathog 7(11):e1002375 10.1371/journal.ppat.1002375 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 296.Kaminski WE, Beham AW, Kzhyshkowska J, Gratchev A, Puellmann K (2013) On the horizon: flexible immune recognition outside lymphocytes. Immunobiology 218(3):418–426. 10.1016/j.imbio.2012.05.024 [DOI] [PubMed] [Google Scholar]
- 297.Mensali N, Dillard P, Hebeisen M, Lorenz S, Theodossiou T, Myhre MR, Fane A, Gaudernack G, Kvalheim G, Myklebust JH, Inderberg EM, Walchli S (2019) NK cells specifically TCR-dressed to kill cancer cells. EBioMedicine 40:106–117. 10.1016/j.ebiom.2019.01.031 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 298.Hu W, Wang G, Huang D, Sui M, Xu Y (2019) Cancer immunotherapy based on natural killer cells: current progress and new opportunities. Front Immunol 10:1205. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 299.Heczey A, Liu D, Tian G, Courtney AN, Wei J, Marinova E, Gao X, Guo L, Yvon E, Hicks J, Liu H, Dotti G, Metelitsa LS (2014) Invariant NKT cells with chimeric antigen receptor provide a novel platform for safe and effective cancer immunotherapy. Blood 124(18):2824–2833. 10.1182/blood-2013-11-541235 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 300.Tosolini M, Pont F, Poupot M, Vergez F, Nicolau-Travers ML, Vermijlen D, Sarry JE, Dieli F, Fournie JJ (2017) Assessment of tumor-infiltrating TCRVgamma9Vdelta2 gammadelta lymphocyte abundance by deconvolution of human cancers microarrays. Oncoimmunology 6(3):e1284723 10.1080/2162402X.2017.1284723 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 301.Deniger DC, Moyes JS, Cooper LJ (2014) Clinical applications of gamma delta T cells with multivalent immunity. Front Immunol 5:636 10.3389/fimmu.2014.00636 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 302.Morita CT, Jin C, Sarikonda G, Wang H (2007) Nonpeptide antigens, presentation mechanisms, and immunological memory of human Vgamma2Vdelta2 T cells: discriminating friend from foe through the recognition of prenyl pyrophosphate antigens. Immunol Rev 215:59–76 [DOI] [PubMed] [Google Scholar]
- 303.Wu YL, Ding YP, Tanaka Y, Shen LW, Wei CH, Minato N, Zhang W (2014) gammadelta T cells and their potential for immunotherapy. Int J Biol Sci 10(2):119–135. 10.7150/ijbs.7823 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 304.Caccamo N, Todaro M, Sireci G, Meraviglia S, Stassi G, Dieli F (2013) Mechanisms underlying lineage commitment and plasticity of human gammadelta T cells. Cell Mol Immunol 10(1):30–34. 10.1038/cmi.2012.42 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 305.Cordova A, Toia F, La Mendola C, Orlando V, Meraviglia S, Rinaldi G, Todaro M, Cicero G, Zichichi L, Donni PL, Caccamo N, Stassi G, Dieli F, Moschella F (2012) Characterization of human gammadelta T lymphocytes infiltrating primary malignant melanomas. PLoS One 7(11):e49878 10.1371/journal.pone.0049878 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 306.Pauza CD, Liou ML, Lahusen T, Xiao L, Lapidus RG, Cairo C, Li H (2018) Gamma delta T cell therapy for cancer: it is good to be local. Front Immunol 9:1305 10.3389/fimmu.2018.01305 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 307.Sheridan BS, Romagnoli PA, Pham QM, Fu HH, Alonzo F III, Schubert WD, Freitag NE, Lefrancois L (2013) gammadelta T cells exhibit multifunctional and protective memory in intestinal tissues. Immunity 39(1):184–195. 10.1016/j.immuni.2013.06.015 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 308.Zhao Y, Niu C, Cui J (2018) Gamma-delta (gammadelta) T cells: friend or foe in cancer development? J Transl Med 16(1):3 10.1186/s12967-017-1378-2 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 309.Lawand M, Dechanet-Merville J, Dieu-Nosjean MC (2017) Key features of gamma-delta T-cell subsets in human diseases and their immunotherapeutic implications. Front Immunol 8:761 10.3389/fimmu.2017.00761 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 310.Handgretinger R, Schilbach K (2018) The potential role of gammadelta T cells after allogeneic HCT for leukemia. Blood 131(10):1063–1072. 10.1182/blood-2017-08-752162 [DOI] [PubMed] [Google Scholar]
- 311.van der Veken LT, Coccoris M, Swart E, Falkenburg JH, Schumacher TN, Heemskerk MH (2009) Alpha beta T cell receptor transfer to gamma delta T cells generates functional effector cells without mixed TCR dimers in vivo. J Immunol 182(1):164–170. 10.4049/jimmunol.182.1.164 [DOI] [PubMed] [Google Scholar]
- 312.Hiasa A, Nishikawa H, Hirayama M, Kitano S, Okamoto S, Chono H, Yu SS, Mineno J, Tanaka Y, Minato N, Kato I, Shiku H (2009) Rapid alphabeta TCR-mediated responses in gammadelta T cells transduced with cancer-specific TCR genes. Gene Ther 16(5):620–628. 10.1038/gt.2009.6 [DOI] [PubMed] [Google Scholar]
- 313.Rischer M, Pscherer S, Duwe S, Vormoor J, Jurgens H, Rossig C (2004) Human gammadelta T cells as mediators of chimaeric-receptor redirected anti-tumour immunity. Br J Haematol 126(4):583–592 [DOI] [PubMed] [Google Scholar]
- 314.Deniger DC, Switzer K, Mi T, Maiti S, Hurton L, Singh H, Huls H, Olivares S, Lee DA, Champlin RE, Cooper LJ (2013) Bispecific T-cells expressing polyclonal repertoire of endogenous gammadelta T-cell receptors and introduced CD19-specific chimeric antigen receptor. Mol Ther 21(3):638–647. 10.1038/mt.2012.267 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 315.Fisher J, Abramowski P, Wisidagamage Don ND, Flutter B, Capsomidis A, Cheung GW, Gustafsson K, Anderson J (2017) Avoidance of on-target off-tumor activation using a co-stimulation-only chimeric antigen receptor. Mol Ther 25(5):1234–1247. 10.1016/j.ymthe.2017.03.002 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 316.van Willigen WW, Bloemendal M, Gerritsen WR, Schreibelt G, de Vries IJM, Bol KF (2018) Dendritic cell cancer therapy: vaccinating the right patient at the right time. Front Immunol 9:2265 10.3389/fimmu.2018.02265 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 317.Braza MS, Klein B (2013) Anti-tumour immunotherapy with Vgamma9Vdelta2 T lymphocytes: from the bench to the bedside. Br J Haematol 160(2):123–132 [DOI] [PubMed] [Google Scholar]
- 318.Hotblack A, Holler A, Piapi A, Ward S, Stauss HJ, Bennett CL (2018) Tumor-resident dendritic cells and macrophages modulate the accumulation of TCR-engineered T cells in melanoma. Mol Ther 26(6):1471–1481. 10.1016/j.ymthe.2018.03.011 [DOI] [PMC free article] [PubMed] [Google Scholar]