Next Article in Journal
Ellagic Acid Controls Cell Proliferation and Induces Apoptosis in Breast Cancer Cells via Inhibition of Cyclin-Dependent Kinase 6
Next Article in Special Issue
Novel Therapies Boosting T Cell Immunity in Epstein Barr Virus-Associated Nasopharyngeal Carcinoma
Previous Article in Journal
MiMIF-2 Effector of Meloidogyne incognita Exhibited Enzyme Activities and Potential Roles in Plant Salicylic Acid Synthesis
Previous Article in Special Issue
Clustering of the ζ-Chain Can Initiate T Cell Receptor Signaling
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

CARs: Beyond T Cells and T Cell-Derived Signaling Domains

1
Department of Dermatology, Universtitätsklinikum Erlangen, Friedrich-Alexander-Universität Erlangen-Nürnberg, Hartmannstraße 14, 91052 Erlangen, Germany
2
Comprehensive Cancer Center Erlangen European Metropolitan Area of Nuremberg (CCC ER-EMN), Östliche Stadtmauerstraße 30, 91054 Erlangen, Germany
3
Deutsches Zentrum Immuntherapie (DZI), Ulmenweg 18, 91054 Erlangen, Germany
*
Author to whom correspondence should be addressed.
These authors contributed equally to this work.
Int. J. Mol. Sci. 2020, 21(10), 3525; https://0-doi-org.brum.beds.ac.uk/10.3390/ijms21103525
Submission received: 29 April 2020 / Revised: 8 May 2020 / Accepted: 13 May 2020 / Published: 15 May 2020
(This article belongs to the Special Issue Recent Advances in T Cell Immunity)

Abstract

:
When optimizing chimeric antigen receptor (CAR) therapy in terms of efficacy, safety, and broadening its application to new malignancies, there are two main clusters of topics to be addressed: the CAR design and the choice of transfected cells. The former focuses on the CAR construct itself. The utilized transmembrane and intracellular domains determine the signaling pathways induced by antigen binding and thereby the cell-specific effector functions triggered. The main part of this review summarizes our understanding of common signaling domains employed in CARs, their interactions among another, and their effects on different cell types. It will, moreover, highlight several less common extracellular and intracellular domains that might permit unique new opportunities. Different antibody-based extracellular antigen-binding domains have been pursued and optimized to strike a balance between specificity, affinity, and toxicity, but these have been reviewed elsewhere. The second cluster of topics is about the cellular vessels expressing the CAR. It is essential to understand the specific attributes of each cell type influencing anti-tumor efficacy, persistence, and safety, and how CAR cells crosstalk with each other and bystander cells. The first part of this review focuses on the progress achieved in adopting different leukocytes for CAR therapy.

1. Conventional T Cells Are the Pioneers of Chimeric Antigen Receptor (CAR) Therapy

T cells are characterized by the possession of a T cell receptor (TCR), in most T cells, consisting of the α and β TCR chains. Mature T cells divide into cell fates defined by the surface co-receptor molecules CD8 (cytotoxic T lymphocytes) and CD4 (T helper and regulatory T cells). Independently of CD4 and CD8, T cells can differentiate from a naïve state (TN) towards an effector (TE) or a memory (TM) phenotype, which is further subdivided in the central memory (TCM) and the effector memory (TEM) compartment, which differ in their self-renewal capacity and effector functions [1,2,3,4,5,6,7].
T cells are clearly the frontrunners of CAR therapy. The first ever CAR created by Gross et al., named T body at that time, was an anti-CD19-CD3ζ CAR (Figure 1) retrovirally transduced into peripheral blood T cells [8]. Over the years, T cells always stayed in the focus of research, with most CAR constructs being designed specifically for this cell type. The greatest success in the CAR field so far and a milestone in cellular therapy was achieved when two autologous anti-CD19-CAR T cell therapies against B cell lymphoma (Kymriah® (Tisagenlecleucel) and Yescarta® (axicabtagen-ciloleucel)) were approved by the Food and Drug Administration (FDA) [9], reaching an astonishing remission rate of 80% [10].
When talking about T cells as CAR vehicles in a generalized way, we must keep in mind that different subpopulations exist. Many published reports did not further differentiate the subtypes and lineages within the expanded T cell pool, meaning that an unknown composition of CD4+, CD8+, naïve, effector, and memory T cells was administered [7]. This becomes important knowing that the frequency of these subsets can differ markedly in individuals because of factors such as age, pathogen exposure, or lymphocytotoxic medications [11,12]. The heterogeneity of T cell subsets may have influenced efficacy and toxicity in clinical trials and could explain part of the variations observed [13,14,15,16], as there are several studies pointing out the influence of the subset distribution on anti-tumor response and persistence [7,17,18,19]. While CD8+ TEM and TCM cells yield the best in vivo persistence of all subsets [20,21], TN and TCM show stronger anti-tumor activity than TEM cells [22,23]. Unfortunately, the TEM subset is usually increased in cancer patients compared to healthy controls [7]. All CD4+ subsets have less cytolytic potential, but show stronger cytokine secretion than CD8+ cells, matching their native role during an immune response [7]. Among both CD4+ and CD8+ T cells, cytokine production is higher in TN than in further differentiated compartments [7]. Sommermeyer et al. determined an ideal cell cocktail to contain 1:1 CD8+ CAR-TCM to CD4+ CAR-TN cells in a mouse model of Raji lymphoma [7], suggesting that IL-2 produced by CD4+ cells drives optimal proliferation of CD8+ CAR-T cells, which are then the main component of anti-tumor cytotoxicity [7,19,24,25]. These findings have been successfully translated to a phase 1/2 clinical trial of an anti-CD19 CAR against acute lymphoblastic leukemia (ALL) (NCT01865617) [26].
Although undoubtedly conventional α/β T cells are the biggest players in the field of CAR cell therapy in the clinics, there are many more cellular vessels to be considered. We will summarize findings with these cell types below.

2. Alternative Cell Types Suitable for CAR Cell Therapy

While having proven their potential in the treatment of hematological cancers [27,28,29,30], CAR therapies have not yet been successfully translated to solid cancers [31,32]. One main hurdle here is the immunosuppressive tumor microenvironment (TME) that impairs recruitment of effector cells and drives them into anergy [33]. Intensifying the treatment, e.g., by increasing the administered dose or by generating more potent CAR effector cells to enhance anti-tumor efficacy, often brings along severe side effects such as cytokine release syndrome (CRS), on-target/off-tumor toxicity, or neurotoxicity [15,16,19,34,35]. This demands for a balance to be found between efficacy and toxicity.
Although α/β T cells remain a very promising vehicle for future CAR therapies, research efforts on other cell types to be used alternatively must be continued. The intrinsic features and unique anti-tumor capabilities of different immune cells might prove better suitable to overcome current hurdles in both efficacy and safety of CAR therapy. This includes abilities to evade suppression by the TME, to drive a broader immune response thanks to cross-interactions among different cell types, to not be restricted to the autologous setting (as conventional α/β T cells are prone to induce graft versus host disease (GvHD), and the reduced tendency towards autoimmunity [36]. Properties and clinical data on the use of less common T cell subsets, NK cells, and myeloid cells as CAR vehicles are summarized in the following.

2.1. γ/δ T Cells

Besides conventional α/β T cells, γ/δ T cells came into focus as potential vehicles for CAR therapy. Their endogenous T cell receptor (although undergoing gene rearrangement) possesses decreased variability and is not restricted to antigens presented on major histocompatibility complex (MHC) [37]. Only some antigens recognized by γ/δ TCRs have been determined so far, e.g., some uncommon nucleotides, phospholipids, and heat shock proteins (HSPs). These are not pathogen-specific themselves but tend to appear on infected or malignant cells under stress, showing the ability of γ/δ TCRs to act as pattern recognition receptors (PPRs) [38]. Additionally, some γ/δ TCRs were shown to bind “non-classical” MHC-Ib molecules and peptides presented on these. Uniting aspects of innate and adoptive immunity, γ/δ T cells are thought to play a “transitional”, “in between” role in the immune system [39,40]. γ/δ T cells are especially prominent in the defense of epithelial tissue, such as the skin or the gut, as part of the population of intraepithelial lymphocytes (IELs) [39]. IEL γ/δ T cells predominantly carry a TCR Vδ1 chain and make up the largest quantity of γ/δ T cells, while a Vδ2 chain carrying subpopulation is circulating in the blood, representing up to 10% of all circulating T lymphocytes [37,41].
γ/δ T cells present within the tumor microenvironment were found to be a favorable diagnostic marker, hinting at these cells’ potential against aberrant cells [37,42]. On the other hand, γ/δ T cells exhibit certain immune regulatory functions in their role as tissue resident cells by secreting the anti-inflammatory cytokines IL-4, IL-10, and TGFβ, which impair effector cells, e.g., within the TME [42]. IL-17, secreted by γ/δ T cells, was even suggested to act tumorigenic by enhancing angiogenesis and the recruitment of macrophages and suppressive myeloid cells [43,44,45,46].
Because they lack affinity for MHC, γ/δ T cells do not exhibit alloreactivity, resulting in several advantages for the use in CAR therapy. γ/δ T cells can be prepared as an ‘off-the-shelf’ therapy when cultured from healthy donor cells [47]. While the patient’s leucocytes might be negatively impacted by previous chemotherapies, donor cells better exhibit their full potential [48]. Furthermore, γ/δ T cells are potentially better suitable to avoid tumor antigen evasion: besides the introduced CAR, they possess a strong anti-tumor potency through their native TCR and various other receptors, including the ability to lyse cells by antibody-dependent cellular cytotoxicity (ADCC) [37]. Blood circulating γ/δ T cells were shown to, under certain conditions, act as antigen-presenting cells (APCs) stimulating conventional α/β T cells [49]. These then launch a poly-antigen targeting attack on the tumor incorporating diverse α/β TCRs [50]. This also allows the buildup of anti-tumor memory, which is key towards long-term tumor control.

Clinical Trials Employing CAR-γ/δ T Cells

Until now, there are three studies listed on clinicaltrials.gov (Table 1), one assessing feasibility of γ/δ T cell isolation and expansion in preparation to testing an anti-CD33-CD28 CAR (Figure 1) (NCT03885076). The other two employ an anti-CD19 CAR against relapsed hematological malignancies (NCT02656147) respectively, an anti-NKG2DL CAR (Figure 1) against a variety of solid tumors (NCT04107142). Neither of them has shared details on their CAR constructs and no results have been published yet.

2.2. NKT Cells

Being less abundant in the blood than γ/δ T cells, NKT cells represent only around 0.1% of circulating T lymphocytes [51]. They co-express NK cell markers and a (semi) invariant α/β TCR, which is restricted to lipid antigens presented on CD1d [51,52]. Proficiency in FasL- and perforin-mediated killing, as well as the cytokine secretion profile, differ between subpopulations defined by CD4, CD8, and CD62L expression [53]. Just like γ/δ T cells, NKT cell invasion in a tumor is considered a favorable predictor for survival in colorectal carcinoma [54], but certain NKT cell subpopulations also have tumor-protective properties preventing immune surveillance [55]. NKT cells possess the ability to recognize and lyse tumor-associated macrophages (TAMs) via their TCR binding CD1d [56], improving tumor invasion and clearing the path for other cell types to follow up. Via CD40L, NKT cells promote the maturation of dendritic cells [57] and thereby the recruiting of α/β T cells to the TME, supporting antigen spread.

Clinical Trials Employing CAR-NKT Cells

An ongoing clinical trial by Heczey and colleagues (NCT03294954, replacing the withdrawn NCT02439788) is testing an (14G2a-mAB-derived) anti-GD2-CD28/IL-15 CAR (Figure 1) construct in autologous NKT cells (called GINAKIT2) (Table 1). Pre-clinical data showed that CD28 co-stimulation and co-expression of IL-15 yielded the best CAR cell durability, relocation to tumor sites, and tumor control in murine neuroblastoma and lymphoma models, while inducing no symptoms of GvHD. Alternatively, 4-1BB co-stimulation (Figure 1) in direct comparison led to reduced proliferation and increased activation-induced cell death.
A second ongoing clinical trial (NCT03774654) also initiated by the Baylor College of Medicine (TX, USA) uses an anti-CD19-CD28/IL-15 CAR construct in allogenic NKT cells against relapsed or refractory B-cell malignancies. The healthy donor-derived transduced CAR-NKT cells (called ANCHOR cells) are administered following immunodepleting therapy. Although previous clinical trials investigated the potential and safety of un-transduced autologous NKT cells in tumor treatment (NCT02619058, NCT01801852), the described two trials represent the first usage of CAR-NKT cells in humans as well as the first clinical use of allogenic NKT cells. To date, no results have been published.

2.3. NK Cells

Natural killer cells exhibit a strong endogenous anti-tumor capacity via a broad variety of natural cytotoxicity receptors (NCRs), stress receptors, and antibody binding Fc(γ) receptors [58], while their cytotoxicity is controlled via inhibitory stimuli from MHC-I binding receptors (e.g., Killer cell immunoglobulin-like receptors, KIRs). NK cells are constantly integrating these activatory and inhibitory signals and enter an activated state once this balance is shifted: upon encountering a cell having downregulated MHC-I (“missing-self”), upon receiving more activatory stimuli from a cell with aberrant surface markers or marked by antibodies (“induced-self”), or a combination of both [58,59]. To lyse tumor cells, NK cells are capable of cellular cytotoxicity and ADCC, as well as secreting cytokines promoting other leucocytes [60]. Two major subpopulations of NK cells have been defined: those residing predominantly in lymphoid organs are CD56hi/CD16low and more proficient in cytokine secretion than in killing [59], while blood circulating NK cells make up around 6% of circulating lymphocytes, are CD56low/CD16hi, and have a higher cytolytic capacity [59].
The lack of alloreactivity allows the usage of heterologous NK cells for adoptive cellular therapy. Until now, most CAR-NK cell studies have stuck to the haplo-identical transplantation setting, although KIR:MHC-I mismatch between graft and recipient was demonstrated to enhance cancer control [61,62]. Clinical scale numbers of heterologous NK cells can be obtained in various ways: by cultivation from PBMCs using autologous PBMCs as feeder cells [63] or IL-15 and 4-1BBL expressing K562 feeders [64], or without any feeder cells [65]. Better persistence of CAR-NK cells was achieved when the cells had been isolated from cord blood [66] or generated by differentiation from gene-modified hematopoietic stem cells [67]. The use of induced pluripotent stem cells has also been described [68]. The NK-92 cell line allows simple and unlimited in vitro expansion of NK cells requiring only IL-2 to grow [69]. Although very convenient to use, this cell line was shown to be limited in its KIR repertoire and lacks CD16 expression, which might render it less effective in tumor control compared to primary NK cells [70]. Furthermore, NK-92 cells are aneuploid and must be irradiated before being administered, limiting their persistence in vivo [70].

Clinical Trials Employing CAR-NK Cells

Besides T cells, NK cells have received the second most attention in CAR research and have been featured in several clinical trials as a potent and save alternative with good pre-sets for future therapeutic application, as defined by pre-clinical data [71,72,73]. Currently (April 2020), a total of seventeen clinical trials employing CAR-transfected NK cells have been registered on clinicaltrials.gov (Table 2).
From all of these, only Liu et al. (NCT03056339) have published results by now [74]. In this phase 1/2 study, heterologous cord blood-derived NK cells have been retrovirally transduced with an anti-CD19-CD28/CD3ζ CAR transcriptionally coupled to IL-15, plus a safety switch in the form of inducible caspase 9 (Figure 1). They were employed against non-Hodgkin’s lymphoma and chronic lymphocytic leukemia (CLL) and achieved a response in eight out of eleven patients, with seven having complete remission. None of the patients showed critically increased levels of inflammatory cytokines or cytokine release syndrome, signs of neurotoxicity, or GvHD. Persistence of the CAR-NK cells was at least twelve months [74]. Of the other ongoing clinical trials, one addresses an anti-BCMA and one an anti-CD3 CAR, and all other trials target CD19. Those who have shared details on their CAR constructs are working on either 4-1BB/CD3ζ or CD28/4-1BB/CD3ζ CARs (Figure 1).

2.4. Myeloid Cells

Beside erythrocytes and platelets, the common myeloid progenitor gives rise to those cells forming the backbone of innate immunity: granulocytes, namely neutrophils, eosinophils, and basophils and mast cells, as well as monocytes, macrophages, and myeloid dendritic cells. Taken together, these cells make up more than half of all blood circulating leukocytes and carry out diverse roles in innate immunity, but also facilitate adaptive immunity.
Very little data on myeloid cells as CAR vehicles has been obtained so far [75,76,77,78] and no clinical trial is registered on clinicaltrials.gov. Other than lymphocytes, the rather short-lived mature granulocytes are difficult to expand in vitro [75]. To obtain therapeutically relevant numbers of CAR-myeloid cells, hematopoietic stem cells (HSCs) or myeloid progenitors can be transduced, (selectively) expanded, and grafted [75]. As a proof-of-principle, De Oliveira et al. retrovirally transduced cord blood-derived human CD34+ hematopoietic stem cells with CD19-specific CAR constructs [75]. After differentiation, the CARs were detectable on mature granulocytes, monocytes, and macrophages without impairing these cells’ conventional effector functions, plus granting them the ability to lyse CD19+ targets in vitro [75]. After transplantation, CAR-expressing neutrophils are expected to appear after about two weeks [36]. The gene-modified HSCs continuously gave rise to new CAR effector cells, including lymphocytes and myeloid cells, resulting in de facto infinite persistence of CAR cells in vivo [76]. While this solves the issue of cell exhaustion faced in conventional CAR-T cell therapy, it also calls for reliable safety switches to control toxicity and the risk of malignant transformation [76]. An example for a safety switch gene successfully tested by Kao et al. is the truncated epidermal growth factor receptor (EGFRt), mediating ADCC [79]. The presence of many different CAR effector cell types at the same time promises multi-level anti-tumor activity, employing all arms of the immune system [77]. Analogously, Hege et al. grafted HSCs transduced with a CD4/CD3ζ fusion construct into mice and measured cytotoxicity against HIV-1 gp120 targets [80]. To demonstrate the contribution of non-T cells to the cytotoxicity, they repeated the experiment in SCID mice (that are incapable generating mature T cells) [80]. Several other groups also proved HSC-derived CAR myeloid cells to significantly contribute to cytotoxicity of this multi-lineage approach [79,81,82]. Of note, most studies working on CAR-transduced HSCs do not address myeloid cells, but focus more on the potential of a self-renewing CAR-T cell response [78].

2.4.1. Neutrophils

Neutrophils are not only the most abundant type of granulocytes but also the most abundant of all leukocytes, making up about 60% to 70% of them [83]. They are very short-lived, having a half-life of only about 18 h in vitro [81]. In an immune response, neutrophils pursue the role of a first line of defense by rapid infiltration of the affected tissue [83]. Only one study, by Roberts et al., has specifically looked at CAR neutrophils so far. The group transduced human HSCs with an anti-CD4-CD3ζ CAR and isolated CAR-expressing neutrophils to successfully test their cytotoxicity against Raji cells in vitro [81].

2.4.2. Monocytes

Monocytes are large-sized leukocytes. Themselves acting as phagocytes in the innate immune system, they can give rise to macrophages and myeloid dendritic cells [84]. Unlike all previously mentioned studies on CAR-myeloid cells, Biglari et al. did not employ gene-modified HSCs but directly isolated peripheral blood human monocytes. They adenovirally transduced the monocytes with an anti-CEA-CD64 CAR (Figure 1) and documented reduced tumor progression in treated mice, accompanied by strong TNF secretion [85].

2.4.3. Macrophages

Macrophages are the most prominent type of phagocyte in the immune system and form several subpopulations with specialized roles. While M1 macrophages are effector cells of pathogen and tumor defense, M2 cells exhibit regulatory and tissue restauration abilities: resolving inflammation and wound healing (M2a), immunoregulation (M2b), immunosuppression and tissue remodeling (M2c), angiogenesis, and also tumor growth (M2d) [86]. This subdivision is not mutually exclusive and mixed phenotypes exist [87]. Macrophages are capable of killing target cells via phagocytosis and trogocytosis (the uptake of severed cell parts) [88,89,90] and MHC-II presentation of antigens taken up, as well as secretion of cytokine and effector molecules [91,92].
Macrophages possess the ability to infiltrate tumors in large numbers and are then referred to as tumor-associated macrophages (TAMs) [93], which can make up 50% of tumor mass [94]. Once in the TME, they lose their capability to migrate and to lyse tumor cells. Adopting an immunoregulatory phenotype, they even contribute to the suppressive TME and tumor growth [33,93,95]. Consequently, the presence of high numbers of TAMs is associated with poor clinical prognosis [93]. On the other hand, increasing the rate of macrophage phagocytosis (e.g., via CD47 blockage) boosts CD8+ T cell responses [96], as the presentation of antigens to T cells is a determining factor of the T cell anti-tumor response [96,97].
The high prevalence of macrophages in solid tumors makes them a promising vehicle for CAR therapy, provided that the suppressive influence of the TME can be overcome [89]. CAR macrophages could contribute to shifting the TME towards tumor rejection by releasing inflammatory cytokines such as TNF, IL-6, IFNγ, and IL-12 [89,98,99], by promoting T cell recruitment and function, and tumor growth suppression [99]. Tumor cell lysis was achieved via CAR-directed phago- and trogo-cytosis [71]. By presenting processed tumor antigens to T cells, CAR macrophages contributed to a poly-antigenic T cell response, reducing the risk of antigen evasion [89,90]. A down-point of macrophages as CAR vehicles is their endogenous resistance to viral infection, resulting in low transduction rates [100]. Adenoviruses yielded the best transduction efficacy but are associated with high immunogenicity and an inflammatory phenotype in target cells, which is a risk factor for clinical applications [89,98,100]. Electroporation was also shown to be inefficient and resulted in low viability [100].
Morrissey et al. lentivirally transduced murine bone marrow-derived macrophages (BMDMs) with anti-CD19 or anti-CD22 CARs using (amongst others) FcRɣ and Megf10 signaling domains (Figure 1) [90]. These so-called chimeric antigen receptors for phagocytosis (CAR-Ps) enabled macrophages to engulf differently sized bead targets and limit Raji cell growth in vitro [90]. Klichinsky et al. employed the THP-1 macrophage model cell line to express anti-CD19-, anti-meso-, and anti-HER2-CD3ζ CARs (CAR macrophages, CARMA) and showed them to selectively clear cognate antigen-bearing tumor cells, promote CTL proliferation, as well as to reduce tumor burden in an ovarian cancer xenograft model [89]. CARMA cells differentiated towards a durable M1 phenotype and resisted subversion to M2. The group also established a chimeric adenoviral vector, Ad5f35, that achieved around 70% transduction efficacy in macrophages [89].
Another group worked on macrophage Toll-like receptor-chimeric antigen receptors (MOTO-CARs), which have a Toll Interleukin receptor (TIR) signaling domain (Figure 1) [98]. Transduced into monocyte-derived human macrophages using Ad5f35, these receptors triggered phagocytosis of non-small cell lung carcinoma cell lines [98].

2.4.4. Myeloid Dendritic Cells

Myeloid dendritic cells (DCs) are antigen-presenting cells (APCs) proficient in antigen uptake, processing, and presentation via MHC molecules. They serve as ambassadors between innate and adaptive immunity, being crucial for antigen-specific T cell responses. While no trial on CAR-transfected dendritic cells has been published so far, cellular vaccination strategies employing DCs are on the rise. Given that a suitable antigen is available, DCs modified to present this particular antigen can drive T cells against virus-infected or cancer cells and initiate multi-lineage immune responses through further cell–cell communications [101]. Immune memory against the designated target is also induced und can grant long-term immunity [101].
As DC vaccination (DCvac) and CAR-T cells are the two fastest growing arms of adoptive cell therapy, recent trials seek to combine these two in the siege against cancer. Wu et al. tested co-cultures of anti-CD19 CAR-T cells with EGFR pathway substrate 8 (Eps8)-presenting DCs in vitro, which enhanced proliferation and reduced AICD of T cells. A shift from the TEM to the favorable TCM compartment increased cytokine secretion (especially IL-2), and lytic capacity was reported when confronted with CD19+/Esp8high leukemia cells [102]. Two clinical trials are registered approaching similar combination therapies (see Table 1). One (NCT03291444) combines a variety of CAR-T cells with Eps8 or WT1 peptide-loaded DCs against ALL, acute myeloid leukemia (AML), and myelodysplastic syndrome. The other (NCT04085159) targets neurofibromatosis with a triple-combination of CAR-T cells, DCvac, and engineered immune effector cytotoxic T cells, without granting more insight on their cells and constructs.
Although not redirecting them directly against target cells, Squadrito et al. presented DCs expressing a chimeric receptor increasing the acquisition of tumor antigens from cancer cell-derived extracellular vesicles (EVs) [103]. Their construct consists of an anti-HER2 single-chain variable fragment (scFv) coupled to a signaling-incompetent truncated nerve growth factor receptor, allowing the DCs to bind EVs carrying MHC-I:HER2-peptide complexes on their surface. Increased internalization of the EVs allowed cross-dressing with the antigen and activation of T cells in vitro [103].
Taken together, since the clinical efficacy of CAR-T cells against solid tumors lags behind their results against hematologic cancers, it is obvious that new approaches are necessary to overcome these limitations. Changing the cellular vehicle encompassing the CAR is one possible way addressed by more and more groups. The strategies listed above will need to prove their efficacy in clinical trials, some of which are currently running. Employing alternative cellular vehicles also demands for specifically optimized CAR constructs, which will be addressed below.

3. Alternative Extracellular and Intracellular Domains Used in CARs

The composition of chimeric antigen receptor constructs has been successively optimized since the first ever CAR was designed in 1989 [8]. The most common CAR design achieves recognition of a target antigen via a monoclonal antibody-derived single chain variable fragment (scFv) [104]. This extracellular domain is connected to intracellular signaling domains via a linker and a transmembrane (TM) region. In the simplest constructs, this signaling domain consists only of the CD3ζ chain from the TCR complex (Figure 1) [8]. Later improvements, referred to as second-generation CARs, added a second signaling domain chosen from the pool of known co-stimulatory molecules, based on the idea that complete T cell activation requires not only “signal 1” via the TCR, but also “signal 2” via co-stimulation (Figure 1) [105,106,107,108]. Different signaling domains grant the CAR cells specific attributes, such as enhanced cytotoxicity, prolonged survival, or stronger proliferation, and increase their overall potency, paving the way to employment against solid tumors [105,106,107,109,110,111,112,113,114,115,116,117,118,119,120].
Building on the success of co-stimulation in second-generation CARs, third-generation receptors have been assembled bearing not one but two or more co-stimulatory domains (Figure 1), expecting even enhanced cell activation, proliferation, and survival [121]. However, experiments showed that the effects of multiple signaling molecules are not contributing in an additive way, but are integrated in a far more complex manner [120]. Results from both preclinical experiments and clinical trials have been published, proving that some third-generation CARs can indeed be superior [73,121,122], while others fall behind [120,121,123] when directly compared to second-generation CARs. This mixed evidence demands for a better understanding of all influencing parameters to allow rational CAR design in the future [120,121,124,125].
Carrying the idea of the “three signals of T cell activation” one step further, CAR cells have been designed to additionally secrete certain cytokines, “signal 3” (Figure 1). These constructs, often revered to as TRUCKs, show the capability to enhance effector functions and durability of effector cells [120,121,126].
Besides the identity and number of employed signaling domains, the design chosen to integrate them into one CAR determines the properties of the receptor as a whole. Sequence and length of the linker and spacer region connecting the scFv domain to the signaling molecules have a strong impact on antigen recognition but also immunogenicity of the entire receptor [19,108,126,127]. The order of intracellular domains in the protein chain and their distance from the plasma membrane greatly impact the signaling competence [120,121,126,128,129,130].
Determining the ideal combination of signaling domains for a certain application is one of the ongoing challenges in the field of CAR therapy, especially when using alternative cell types. The following sections summarize properties and experimental data on the classical as well as uprising alternative signaling domains used in CARs over the last years.

3.1. Classical Signaling Domains

CD3ζ, CD28, and 4-1BB (i.e., CD137) today are the best studied signaling domains used in CAR constructs across different cell types. Years of pre-clinical testing with such constructs have led to the FDA approval of the first two CAR-T cell therapies in 2017 [9]: Kymriah and Yescarta, both directed against CD19 but containing different TM and signaling domains (CD8α(TM)-4-1BB/CD3ζ respectively IgG1(TM)-CD28/CD3ζ) [9], clearly making them the hallmarks of CAR research.

3.1.1. CD3ζ

Two CD3ζ chains, also called CD247, are part of the native T cell receptor complex. Together with one CD3γ and -δ chain and two CD3ε chains, they form an open barrel structure around a TCR α/β dimer respectively, a TCR γ/δ dimer, interacting via residues within the membrane interface [131,132] (Figure 2). In its intracellular part, each CD3ζ chain contains three tyrosine-rich sequences known as immunoreceptor tyrosine-based activation motifs (ITAMs) [133]. These are phosphorylated by membrane-localized Src-kinases upon TCR antigen binding, predominantly by Lyn, which associates with the MHC-binding co-receptors CD4 and CD8 to be brought in proximity of the TCR complex [134]. Phosphorylation renders ITAMs capable of binding the kinase ZAP70, leading to down-stream signaling via the MAPK, NF-κB, and NF-AT pathways in T cells. By promoting the transcription of corresponding genes, these pathways promote survival, proliferation, maturation, secretion of IL-2 and other cytokines, and cytolytic functions [135].
Although CD3ζ is only expressed on T and NK cells, signaling cascades involving ITAMs are present in all leucocytes, meaning that phosphorylation of CD3ζ can induce down-stream effects in different cell types. While the ITAM binding kinase ZAP70 is only expressed in T and NK cells, the closely related kinase Syk is present in all non-T leucocytes. In NK cells, neutrophils, macrophages, and mast cells, ITAM phosphorylation mimics Fc-receptor signaling and therefore triggers ADCC respectively to uptake of antibody-coated targets.
In first-generation CARs (Figure 1), the transmembrane domain of CD3ζ was used to connect the signaling domain to the extracellular scFv domain [8]. As the CD3ζ TM domain contains those residues essential for the association with the endogenous TCR complex, it was shown that these CARs have the ability to cluster with endogenous TCRs on the T cell surface [136]. In second- and third-generation CARs, it is very uncommon to use the CD3ζ TM domain, and most constructs rely on the CD28- or CD8α-TM domain [126,137].

3.1.2. CD28

CD28 is a receptor expressed on T cells acting as a co-stimulatory signal supporting the TCR (Figure 2). Besides on T cells, CD28 is also expressed on eosinophils and neutrophils [138,139,140]. Its ligands are CD80 and CD86, present on antigen-presenting cells. As a co-stimulatory molecule, its absence or presence on antigen-presenting cells plays an important role in peripheral tolerance because naïve T cells can only become activated if the TCR and CD28 are simultaneously stimulated. When a T cell is interacting with an antigen-presenting cell, CD28 is a major driver in immunological synapse organization [141] by manipulating the actin cytoskeleton [142]. This rearrangement of the cytoskeleton also guarantees recruitment of binding partners in close proximity to the TCR complex and, for example, facilitates ITAM phosphorylation. In this manner, CD28 co-stimulation supports the MAPK, NF-κB, and NF-AT pathways, enhancing all effector functions stimulated by TCR signaling [143]. On a functional level, CD28 promotes the production of IL-2, -6, -10, and further interleukins, as well as cell cycle progression, survival, differentiation, and cytolytic functions [144].
A long list of studies employing CARs with a CD28 signaling domain reported potent and quick anti-tumor effector functions but found these to be rather short-lived and associated with limited cell persistence in vivo when compared to, e.g., the 4-1BB signaling domain [113,143,145,146]. CD28 co-stimulated CAR-T cells differentiate towards an effector-memory type which is characterized by secretion of IL-2 and IL-10, and an upregulated glycolytic metabolism to sustain their high energy demand [27,113,121,147,148,149,150,151,152]. In many second- and third-generation CAR constructs, the TM domain of CD28 is used as a connector of extra- and intra-cellular domains, which was associated with improved expression and presence of these CARs on the surface [153,154]. CD28 co-stimulation (potentially enhanced by the high expression levels) is thought to facilitate tonic CAR signaling [106,155] and thereby lead to Fas-dependent activation-induced cell death (AICD) in CAR-T cells [114]. This might also explain the observed limited cell persistence [113]. Clinical data mirrors preclinical findings: CD28 supports strong but short-lived anti-tumor efficacy [16,156,157]. Although CD28 is one of the most commonly employed signaling domains in CAR-T cells, it has been questioned whether CD28 can successfully enhance activation in CAR-NK cells [126], since NK cells lack naturally expressed CD28 [158,159].

3.1.3. 4-1BB (CD137)

4-1BB is known as a co-stimulatory receptor that, together with others, is upregulated on the surface of activated T cells and promotes prolonged activation (Figure 2). Apart from activated T cells, it is expressed on a variety of lymphoid cells [160,161] as well as some non-hematopoietic tissues [162,163]. 4-1BB signaling in T cells enhances cell cycle progression and proliferation, cytokine secretion, cytolytic potential, and inhibits clonal deletion and AICD [164,165].
When employed as a CAR signaling domain (Figure 1), 4-1BB was shown to increase cytotoxicity above the level of first generation CARs and allow for more robust cell activation, as well as increase persistence in vivo [27,107,121,145,147,148,149,150,151,152,166]. By altering the cell’s metabolism towards enhanced respiratory capacity and fatty acid oxidation, 4-1BB co-stimulation promotes differentiation of CAR-T cells towards a central memory phenotype [27,113,121,147,148,149,150,151,152]. When compared to CD28, 4-1BB co-stimulated CARs showed slower onset of cytotoxicity, but longer durability and accumulation of CAR cells over time, which ultimately resulted in comparable efficacy but slower kinetics of the anti-tumor response [107].
Clinical trials similarly reported potent anti-tumor efficacy and very long persistence of 4-1BB-containing CAR-T cells in patients, with transgenic cells being detectable for up to several years [167,168]. Good in vivo persistence is a prerequisite to build up anti-tumor memory and long-term tumor control [107,167,168].

3.2. Non-Classical Extracellular and Signaling Domains

CAR constructs employing CD3ζ plus either CD28, 4-1BB, or both, have become state-of-the-art in laboratory work and clinical trials. To further develop CAR therapy regarding efficacy and safety, to direct it towards solid cancer types, and to bring new cellular vehicles into play, the optimization of the signaling capacity of CARs needs to be continued. Employing new signaling domains will reveal especially effective combinations among them and show specific potencies in different cell types. The following sections summarize properties and reported trials of a selection of proteins that are or might become emerging candidates for next generation CARs.

3.2.1. NKG2D

NKG2D is a C-type lectin-like receptor expressed by NK, CD8+ α/β T, and γ/δ T cells (Figure 2) [27,169,170]. It is activated by induced self-proteins known as stress markers on aberrant, such as infected or malignant, cells [27,171,172,173,174]. The most prominent role of NKG2D is to drive NK cell-mediated cellular cytotoxicity against tumors [175]. In cytotoxic CD8+ T lymphocytes, the receptor delivers co-stimulation to TCR signaling [170]. Two NKG2D proteins form a dimer and each monomer complexes with one DAP10 dimer for down-stream signaling (Figure 2) [54,55,176,177], which is addressed in more detail below.
Being limited to just one antigen has ever since been the weak spot of scFv-based CARs, restricting their field of application to cancer types with known characterizing antigens, and bearing the risk of tumor antigen evasion. Several groups worked on a different approach on chimeric receptors, employing a complete NKG2D receptor molecule fused with the inverted intracellular domain of CD3ζ, called NKR2 (Figure 1) [178]. Instead of recognizing a specific antigen, this CAR’s extracellular domain binds stress markers just as the endogenous receptor does. The CAR intracellularly complexes with the adapter protein DAP10 for co-stimulatory signaling [55,176,177,179], while the CD3ζ domain drives cell activation and cytotoxicity [178]. Not being limited to one target antigen makes this CAR a promising tool against different cancer types and outplays the risk of tumor antigen evasion [178,180]. Preclinical data shows the NKR2 to be potent against a variety of hematologic and solid cancers [178,181,182,183]. NKR2 T cells have been translated to six phase 1/2 clinical trials (NCT02203825, NCT03018405, NCT03310008, NCT03370198, NCT03466320, and NCT03692429), employed against acute myeloid leukemia (AML), multiple myeloma (MM), melodysplastic syndrome, colorectal cancer, and colon cancer liver metastases.
A slightly different chimeric NKG2D receptor combines NKG2D with CD28 and CD3ζ signaling molecules [184]. Lentivirally transduced and mRNA-transfected T cells were tested against Ewing’s sarcoma family of tumors. Tumor cells were efficiently lysed by both CD8+ and CD4+ T cells, although the receptor disappeared from the surface of transiently transfected cells after a few hours [184].
While all of these CARs included the extracellular domain of the NKG2D receptor, Li et al. presented a CAR consisting of an anti-mesothelin (meso) scFv coupled to only the transmembrane domain of NKG2D plus 2B4 (see below) and CD3ζ signaling domains (Figure 1) [68]. The group pointed out that many previous studies of CAR-NK cells translated CAR constructs originally designed for the use in T cells to NK cells, although signaling domains result in different downstream effects dependent on the cell type. Adequate CAR constructs have to be chosen to unleash a vehicles full potential [68]. The group generated and screened nine CARs employing only domains with prominent roles in NK cells, chose the most promising candidate (anti-meso-NKG2D/2B4/CD3ζ), and went on to further characterizing it. NK cells derived from transduced induced pluripotent stem cell (iPSC) effectively inhibited cancer growth and prolonged survival in meso+ ovarian cancer-xenografted mice, outperforming iPSC NK cells expressing a pre-known CD28/4-1BB/CD3ζ CAR. When comparing the novel NKG2D/2B4/CD3ζ CAR-NK cells to T cells transduced with the CD28/4-1BB/CD3ζ CAR, both demonstrated very similar anti-tumor activity, fortifying the importance of vehicle-specific CAR design. In terms of persistence, the new NK-cell CAR was superior to its rival and resulted in less off-target toxicity. When tracking down the effector functions to the individual domains of the CAR via selective introduction of disruptive mutations, it became clear that the NKG2D and 2B4 domains both significantly contributed to activatory signaling, while CD3ζ only played a minor role in this construct [115].

3.2.2. DAP10

An NKG2D dimeric receptor associates with two dimers of the adaptor protein called DNAX-activating protein of 10 kDa (DAP10) via their TM domains to form a hexameric complex (Figure 2) [54,55,176,177]. Upon antigen binding, the cytoplasmic domain of DAP10 becomes phosphorylated and recruits PI3K and a complex of GRB2 and VAV1 to trigger down-stream signaling cascades [54,55,176,177].
Comparing the cytokine profile of different CAR cells, secreted cytokines usually include pro-inflammatory IFNγ, TNF, IL-2, GM-CSF, IL-17, and IL-21, as well as anti-inflammatory IL-10 [152]. Unlike most other signaling domains, DAP10 was shown not to induce IL-10 secretion, but to strongly enhance T cell effector functions via pro-inflammatory cytokines [152,182,185,186,187,188,189,190,191,192,193]. Similar to CD28-bearing receptors, DAP10 promoted differentiation towards an effector memory phenotype in T cells, which was associated with enhanced persistence in vivo [27,121,147,148,149,150,151,152,182,185,186,187,188,189,190,191,192].
Several groups have performed studies on novel CAR constructs bearing a DAP10 signaling domain over the last years. Their results are summarized in the following. When elongating two second-generation CD28/4-1BB/CD3ζ CARs by a DAP10 domain, Zhao et al. found these receptors to have superior anti-tumor activity against GPC3+ lung cancer, respectively MSLN+ hepatocellular carcinoma and gastric cancer, prolonging the survival of xenograft-bearing mice [194,195].
In their proof-of-principle study, Duong et al. screened a variety of randomly assembled CARs for potent constructs. By this, they brought up a promising DAP10/CD3ζ/CD27 CAR and probed it in T cells versus ErbB2+ tumor cells, both in vitro and in a mouse model. They reported the new CAR to perform significantly better than an established CD28/CD3ζ CAR [196].
CARs based on the extracellular binding domain of PD-1 can be employed against a broad variety of cancer types that upregulate PD-1 ligands on their surface. When testing PD1-CD3ζ/DAP10 CAR-T cells in a mouse model of lymphoma, Lynch et al. saw significantly enhanced persistence compared to a CD28 co-stimulated CAR, leading to long-term tumor-free survival [192]. PD1-DAP10/CD3ζ CAR-T cells employed against syngeneic murine models of a variety of solid cancers (renal, pancreatic, liver, colon, breast, prostate, and bladder cancer, and melanoma) were also capable of inducing remission [193]. Both groups accredited the good performance to the preferential (IL-10-free) cytokine profile and memory phenotype differentiation induced by the DAP10 domain [192,193].
Two hurdles faced in CAR therapy that both result from powerful cell activation domains, are, on one side, on-target/off-tumor toxicity, and on the other side, tonic signaling, resulting in effector cell exhaustion. In an attempt to address both of these issues, Fisher et al. tested a “co-stimulation only” CAR (chimeric co-stimulation receptor, CCR), consisting of an anti-GD2 extracellular domain and DAP10 as the sole signaling domain (Figure 1), transduced in γ/δ T cells [197,198]. Because the DAP10 signal alone is not sufficient for activation, these T cells depend on simultaneous activation of their native γ/δ TCR [197]. In vitro, the CCR T cells efficiently lysed neuroblastoma cells but spared GD2+ cells lacking γ/δ TCR ligands [197]. CCR T cells also differentiated between healthy and malignant myeloid target cells [198], making chimeric co-stimulation receptors a very safe potential alternative to established CARs.
While the NKR2 receptor described in the chapter above consisted only of NKG2D and a CD3ζ signaling domain, Chang et al. designed a CAR additionally bearing the DAP10 domain (Figure 1) in both retrovirally transduced and mRNA-electroporated NK cells [180]. The CAR cells showed potent anti-tumor activity in a mouse model of osteosarcoma which is unresponsive to native NK cells, but no comparison to other receptors was performed [180].
Two trials have been conducted employing NK cells, respectively cytokine-induced killer cells transduced with anti-CD19 first- and second-generation CARs bearing a DAP10 signaling domain (Figure 1) [166,199]. Unlike the ones listed above, these trials yielded dampening results: although DAP10 signaling is a strong trigger of cytotoxicity in native NK cells [200,201], those CARs induced weaker anti-tumor activity in direct comparison to receptors containing CD3ζ alone, respectively 4-1BB/CD3ζ or CD28/CD3ζ [166,202]. These findings stand in clear contrast to others listed above. A better understanding of all factors influencing the potency of these particular CAR constructs is needed to draw reliable conclusions.

3.2.3. Fcγ-receptors

By binding to the Fc part of antibodies, Fc-receptors induce immune cell effector functions towards antibody-bound targets (Figure 2) [203,204,205]. They drive phagocytosis of antibody-opsonized extracellular pathogens by macrophages, dendritic cells, and neutrophils, as well as ADCC by NK cells, eosinophils, basophils, and mast cells via exocytosis of cytotoxic mediators [203,204,205]. Besides these activatory functions, some Fc-receptors also pursue regulatory roles, facilitate antigen uptake and presentation, and allow placental transport of antibodies [206]. Receptors in this family differ in their specificity and affinity for certain antibody classes and subclasses, and are selectively expressed by different leucocytes [204]. Due to their strong activatory properties in many cell types, Fc-receptors are an interesting group of proteins to be employed in CARs.

FcγRIII (CD16)

The best-studied Fc-receptor is the type III Fcγ receptor (FcγRIII), also known as CD16 (Figure 2), which binds IgG1 and 3 with moderate affinity [203,207,208,209]. Two subtypes of this receptor, A and B, sharing 96% sequence identity [210], are characteristically expressed on certain cell types: type A plays the most prominent role by guiding NK cells towards antibody-opsonized cells and inducing ADCC—leading to the release of perforin and granzymes from granules. It also stimulates cytokine release in macrophages [207,208]. Type B initiates exocytosis of granules, mainly against microbes in eosinophils, macrophages, neutrophils, and mast cells [207,208]. To initiate down-stream signaling, FcγRIIIA associates with a dimer of the Fcγ adaptor protein, which is closely related to CD3ζ, or a CD3ζ dimer, or a heterodimer of the two (Figure 2), depending on the native expression of these proteins is the respective cell [209]. Phosphorylation of ITAMs on these adapter proteins initiates the next steps in the signaling cascade [207,208].
Different strategies have been tested to develop CARs not limited to just one tumor antigen. Universal CARs (uniCARs) do not have a covalently linked scFv but can reversibly complex with antibodies. One way to implement this is to generate Fcγ chimeric receptors (Fcγ CRs) by coupling intracellular co-stimulatory plus signaling domains to an extracellular CD16 domain, which then binds to the Fc part of antibodies [211,212]. Fcγ CRs can synergize with antibodies naturally present in the patient and be redirected to specific target antigens by administration of therapeutic monoclonal antibodies (mAbs) [211,212]. In contrast to scFv-based CARs, this allows to employ Fcγ CR cells for a broad variety of malignancies for which therapeutic mAbs are available, and to fine-tune the therapy for the individual patient via frequency and dosage [211,212]. The usage of a cocktail of different mAbs reduces the risk of antigen evasion and allows to react to uprising tumor cells with an altered phenotype [211,212]. In terms of safety, the therapy can be adjusted to control toxicity and, if required, quickly be interrupted by ending mAb administration [211,212].
Fcγ CRs have been pre-clinically tested by different groups [213,214,215,216,217,218,219]. Summarizing these studies, CD16A or a high-affinity variant (V158) was either coupled directly or via a CD8α linker and TM domain to a signaling tail, consisting of CD3ζ, 4-1BB/CD3ζ, or FcεRIγ (Figure 1). These constructs have been retrovirally transduced or transfected [214,215] into human T cells, murine MD45 T cells, human peripheral blood NK cells, or NK-92 cells. These cells have been employed in combination with a variety of mAbs (anti-CD19, anti-CD20 (Rituximab), anti-HER2 (Trastuzumab), anti-GD2, anti-Panc1, anti-EGFR (Cetuximab, Panitumumab), or anti-CSPG4) against different cancer cells (Daudi, Raji, and adult T cell lymphoma, neuroblastoma, osteosarcoma, pancreatic cancer, colorectal cancer, and A375 melanoma cells) in vitro, as well as against lymphoma-xenografted mice [213,214,215,216,217,218,219]. Fcγ CR cells showed promising anti-tumor activity in all these studies [213,214,215,216,217,218,219], and via the attached signaling domains, activated the NF-AT pathway [213]. Fcγ CR T cells expressing the 4-1BB/CD3ζ signaling domains sustained especially potent proliferation, and stronger activation and cytotoxicity than with CD3ζ or FcεRIγ [214]. Fcγ CR T cells preferentially differentiated towards the favorable effector memory phenotype [213]. In direct comparison, Fcγ CR NK cells achieved comparable ADCC, but showed less proliferation than Fcγ CR T cells, causing decreased persistence and reduced long-term tumor control [213].
Fcγ CRs were the first implementation of uniCARs to reach the clinical phase [212]. A total of three phase 1 and one phase 1/2 trials have been registered (clinicaltrials.gov: NCT02315118, NCT02776813, NCT03266692, and NCT03189836), three employing CD16(V158)-4-1BB/CD3ζ autologous T cells in combination with Rituximab against refractory or relapsed lymphoma, respectively with BCMA against relapsed or refractory multiple myeloma. The fourth trial also targets refractory or relapsed lymphoma using Rituximab and an unknown construct in T cells.

FcγRIIA (CD32A)

The type II Fcγ receptor subtype A (FcyRIIA) (Figure 2) binds all subtypes of IgG with moderate affinity. It is expressed on macrophages and neutrophils, where it facilitates phagocytosis, and, on eosinophils, where it triggers degranulation. Besides that, the receptor is found on platelets and Langerhans cells. Unlike FcγRIII, FcγRIIA does not need to associate with Fcγ or CD3ζ for signaling. It possesses an Fcγ-like intracellular domain containing one ITAM that can be phosphorylated by Src kinases (Figure 2).
Based on the emergence of Fcγ CRs using a CD16 extracellular domain, Caratelli et al. designed a CD32A131R (low-affinity variant) chimeric receptor with a CD8α TM and a CD28/CD3ζ signaling domain to be retrovirally transduced into peripheral blood T cells [220]. Contrary to CD16, CD32A has moderate affinity also for IgG2, which is important in the treatment of epidermal growth factor receptor (EGFR)-positive tumors: beside Cetuximab (IgG1), Panitumumab (IgG2) is used as a therapeutic antibody. Directly comparing CD32A CR- to CD16 CR-T cells in combination with both those antibodies in vitro showed superior lytic capacity of the former construct, benefiting from both mAbs directing ADCC, while CD16 can only bind one of them [220].
The group went on translating their approach to KRAS-mutated colorectal cancer (CRC), which evades NK cell-mediated ADCC when treated only with Cetuximab and Panitumumab [219]. The previously presented CD32A131R CD8αTM/CD28/CD3ζ CR was compared to CD32131H, CD16158F, and CD16158V CRs. In this setting, CD32 CR-T cells failed to produce pro-inflammatory cytokines or exhibited cytotoxicity when co-cultured with opsonized KRAS-mutated HCT116 cells, although successfully binding the mAbs, while the CD16158V CR was found potent both in vitro and in vivo [219].

3.2.4. 2B4 (CD244)

The signaling lymphocyte activation molecule (SLAM)-related receptor 2B4 (Figure 2) is a non-MHC-restricted immune regulatory trans-membrane receptor expressed predominantly on NK cells and some T cells (NKT, γ/δ T, and a portion of CD8+ T cells), but also on eosinophils, basophils, mast cells, monocytes, and dendritic cells [221,222]. Together with other SLAM family proteins, it contributes to the formation of the immunological synapse in NK and T cells, on one hand modulating cellular cytotoxicity, mediating a potent (co-)stimulatory signal, or on the other hand, driving cells into exhaustion; depending on multiple, not yet entirely understood factors [221,222,223,224,225,226]. Binding of its ligand, CD48, triggers phosphorylation of immunoreceptor tyrosine-based switch motifs (ITSMs) in the cytoplasmic tail of 2B4 (Figure 2) [115]. Of the four ITSMs present, only the first and second are associated with activatory signaling in NK cells [226]. When driving an activatory response, 2B4 is thought to act mainly as a co-stimulatory receptor crosslinking with, e.g., CD226 or NKG2D, and activating PLC-γ2, triggering Ca2+ influx, stimulating NK cell degranulation, proliferation, IL-2 secretion, and IFNγ release [63,222,226].
It has been shown that 2B4 expression is altered under specific pathologic conditions and might contribute to immune suppression in the TME [222]. Inhibitory signaling of 2B4 was predominantly found in chronic infection or in tumor-associated NK and T cells, seemingly being dependent on surface density of 2B4 and SLAM-associated protein (SAP), a binding partner of CD48: a high ratio of 2B4 to SAP correlates with inhibitory signaling, a low ratio with activatory signaling [222]. As these findings are mainly derived from NK cell studies, little is known about the role of 2B4 and its divergently expressed binding partners in other cell types [222].
2B4 has been studied as a signaling domain in CAR-NK cells by Altvater et al.: a CD19-specific scFv coupled to CD3ζ or 2B4 alone, 2B4/CD3ζ, or 4-1BB/CD3ζ signaling domains were tested against the leukemic cell line REH in vitro [225]. 2B4 was truncated to remove the latter two ITSMs which might drive inhibitory signaling [225]. 2B4 alone yielded a CAR that showed very weak activation, being less potent than a CD3ζ CAR, but combining 2B4 and CD3ζ performed similarly well to 4-1BB/CD3ζ [225,226]. Building on these findings, anti-GD2-2B4/CD3ζ CAR-NK cells were successfully employed against neuroblastoma cells in vitro [225]. To survey the effects of 2B4 signaling on T cells, the anti-CD19, respectively anti-GD2-2B4/CD3ζ CARs were transduced into peripheral blood T cells, yielding results comparable to NK cells, as presented above [226]. Proliferation of these T cells predominantly gave rise to the effector memory subset, which is associated with sustained strong anti-tumor capacity. The tumor-protective Treg subset was not expanded, supporting the role of 2B4 as a co-stimulatory receptor in T cells [226].
As described above in more detail, Li et al. designed a novel CAR specifically for the use in NK cells [68]. Anti-meso-NKG2D/2B4/CD3ζ CAR iPSC NK cells outperformed NK cells expressing an established CD28/4-1BB/CD3ζ CAR. It also had an edge over T cells expressing this classical CAR, when tested in meso+ ovarian cancer xenografted mice. NKG2D and 2B4 were both shown to contribute to the CAR’s performance [68].
In another study aiming to prove the importance of vehicle-specific CAR design, two anti-CD5 CARs with a 4-1BB/CD3ζ (a T cell-associated) or a 2B4/CD3ζ (rather NK cell-specific) intracellular tail were tested [227]. Both were transduced into NK cells and employed in a CD5+ T cell acute lymphoblastic leukemia (T-ALL) mouse model, where they both exhibited specific cytotoxicity and prolonged survival of the mice. The 2B4/CD3ζ CAR displayed greater lytic capacity but also elevated toxicity against non-malignant T cells [227].
In summary, the above shows that many alternative extracellular domains and intracellular signaling modules besides the classical scFv, CD3ζ, CD28, or 4-1BB domains are already tested for functionality in CARs. It is pivotal, however, to find ideal combinations between cellular vessels and CARs with different signaling moieties.

Author Contributions

Conceptualization, N.M.S., J.D., and N.S.; investigation, N.M.S., J.D., and N.S.; writing—original draft preparation, N.M.S.; writing—review and editing, N.M.S., J.D., and N.S.; visualization, N.M.S. and N.S. All authors have read and agreed to the published version of the manuscript.

Funding

This research was funded by the Deutsche Forschungsgemeinschaft (German Research Foundation), trilateral grant SCHA 1247/3-1.

Acknowledgments

We acknowledge support by the Friedrich-Alexander-Universität Erlangen-Nürnberg (FAU) within the funding program Open Access Publishing.

Conflicts of Interest

The authors declare no conflict of interest.

Abbreviations

CARChimeric antigen receptor
TCRT cell receptor
MHCMajor histocompatibility complex
APCAntigen-presenting cell
TEEffector T cell
TMMemory T cell
TCMCentral memory T cell
TEMEffector memory T cell
ALLAcute lymphoblastic leukemia
TMETumor microenvironment
CRSCytokine release syndrome
GvHDGraft versus host disease
HSPHeat shock protein
PPRPattern recognition receptor
IELIntraepithelial lymphocyte
ADCCAntibody-dependent cellular cytotoxicity
NCRNatural cytotoxicity receptor
KIRKiller cell immunoglobulin-like receptor
CLLChronic lymphocytic leukemia
HSCHematopoietic stem cell
EGFREpidermal growth factor receptor
TAMTumor-associated macrophage
BMDMBone marrow-derived macrophage
CAR-PChimeric antigen receptor for phagocytosis
CARMACAR macrophage
TIRToll interleukin receptor
DCDendritic cell
DCvacDC vaccination
Eps8EGFR pathway substrate 8
EVExtracellular vesicle
scFvSingle-chain variable fragment
TMTransmembrane
ITAMImmunoreceptor tyrosine-based activation motif
AICDActivation-induced cell death
AMLAcute myeloid leukemia
MMMultiple myeloma
mesoMesothelin
iPSCInduced pluripotent stem cell
DAP10DNAX-activating protein of 10 kDa
CCRChimeric co-stimulatory receptor
uniCARUniversal CAR
Fcγ CRFcγ chimeric receptor
mAbMonoclonal antibody
EGFREpidermal growth factor receptor
CRCColorectal cancer
SLAMSignaling lymphocyte activation molecule
ITSMImmunoreceptor tyrosine-based switch motif
SAPSLAM-associated protein
T-ALLT cell acute lymphoblastic leukemia

References

  1. Kaech, S.M.; Wherry, E.J.; Ahmed, R. Effector and memory T-cell differentiation: Implications for vaccine development. Nat. Rev. Immunol. 2002, 2, 251–262. [Google Scholar] [CrossRef] [PubMed]
  2. Stemberger, C.; Neuenhahn, M.; Buchholz, V.R.; Busch, D.H. Origin of CD8+ effector and memory T cell subsets. Cell. Mol. Immunol. 2007, 4, 399–405. [Google Scholar] [PubMed]
  3. Gattinoni, L.; Lugli, E.; Ji, Y.; Pós, Z.; Paulos, C.M.; Quigley, M.F.; De Almeida, J.R.; Gostick, E.; Yu, Z.; Carpenito, C.; et al. A human memory T cell subset with stem cell–like properties. Nat. Med. 2011, 17, 1290–1297. [Google Scholar] [CrossRef]
  4. Gerlach, C.; Rohr, J.C.; Perie, L.; Van Rooij, N.; Van Heijst, J.W.J.; Velds, A.; Urbanus, J.; Naik, S.H.; Jacobs, H.; Beltman, J.; et al. Heterogeneous Differentiation Patterns of Individual CD8+ T Cells. Science 2013, 340, 635–639. [Google Scholar] [CrossRef] [PubMed]
  5. Buchholz, V.R.; Flossdorf, M.; Hensel, I.; Kretschmer, L.; Weissbrich, B.; Gräf, P.; Verschoor, A.; Schiemann, M.; Höfer, T.; Busch, D.H. Disparate Individual Fates Compose Robust CD8+ T Cell Immunity. Science 2013, 340, 630–635. [Google Scholar] [CrossRef] [PubMed]
  6. Graef, P.; Buchholz, V.R.; Stemberger, C.; Flossdorf, M.; Henkel, L.; Schiemann, M.; Drexler, I.; Höfer, T.; Riddell, S.R.; Busch, D.H. Serial Transfer of Single-Cell-Derived Immunocompetence Reveals Stemness of CD8+ Central Memory T Cells. Immunity 2014, 41, 116–126. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  7. Sommermeyer, D.; Hudecek, M.; Kosasih, P.L.; Gogishvili, T.; Maloney, D.G.; Turtle, C.J.; Riddell, S.R. Chimeric antigen receptor-modified T cells derived from defined CD8+ and CD4+ subsets confer superior antitumor reactivity in vivo. Leukemia 2015, 30, 492–500. [Google Scholar] [CrossRef] [Green Version]
  8. Gross, G.; Gorochov, G.; Waks, T.; Eshhar, Z. Generation of effector T cells expressing chimeric T cell receptor with antibody type-specificity. Transplant. Proc. 1989, 21, 127–130. [Google Scholar]
  9. FDA Approves Second CAR T-cell Therapy. Cancer Discov. 2017, 8, 5–6. [CrossRef] [Green Version]
  10. Yip, A.; Webster, R.M. The market for chimeric antigen receptor T cell therapies. Nat. Rev. Drug Discov. 2018, 17, 161–162. [Google Scholar] [CrossRef]
  11. Mackall, C.L.; Fleisher, T.A.; Brown, M.R.; Andrich, M.P.; Chen, C.C.; Feuerstein, I.M.; Horowitz, M.E.; Magrath, I.T.; Shad, A.T.; Steinberg, S.M.; et al. Age, Thymopoiesis, and CD4+ T-Lymphocyte Regeneration after Intensive Chemotherapy. N. Engl. J. Med. 1995, 332, 143–149. [Google Scholar] [CrossRef] [PubMed]
  12. Hakim, F.T.; Cepeda, R.; Kaimei, S.; Mackall, C.L.; McAtee, N.; Zujewski, J.; Cowan, K.; Gress, R.E. Constraints on CD4 Recovery Postchemotherapy in Adults: Thymic Insufficiency and Apoptotic Decline of Expanded Peripheral CD4 Cells. Blood 1997, 90, 3789–3798. [Google Scholar] [CrossRef] [PubMed]
  13. Brentjens, R.J.; Rivière, I.; Park, J.H.; Davila, M.L.; Wang, X.; Stefanski, J.; Taylor, C.; Yeh, R.; Bartido, S.; Borquez-Ojeda, O.; et al. Safety and persistence of adoptively transferred autologous CD19-targeted T cells in patients with relapsed or chemotherapy refractory B-cell leukemias. Blood 2011, 118, 4817–4828. [Google Scholar] [CrossRef] [PubMed]
  14. Kalos, M.; Levine, B.L.; Porter, D.L.; Katz, S.I.; Grupp, S.A.; Bagg, A.; June, C.H. T Cells with Chimeric Antigen Receptors Have Potent Antitumor Effects and Can Establish Memory in Patients with Advanced Leukemia. Sci. Transl. Med. 2011, 3, 95ra73. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  15. Kochenderfer, J.N.; Dudley, M.E.; Feldman, S.A.; Wilson, W.H.; Spaner, D.E.; Maric, I.; Stetler-Stevenson, M.; Phan, G.Q.; Hughes, M.S.; Sherry, R.M.; et al. B-cell depletion and remissions of malignancy along with cytokine-associated toxicity in a clinical trial of anti-CD19 chimeric-antigen-receptor–transduced T cells. Blood 2012, 119, 2709–2720. [Google Scholar] [CrossRef]
  16. Davila, M.L.; Bouhassira, D.C.G.; Park, J.H.; Curran, K.J.; Smith, E.L.; Pegram, H.J.; Brentjens, R.J. Chimeric antigen receptors for the adoptive T cell therapy of hematologic malignancies. Int. J. Hematol. 2013, 99, 361–371. [Google Scholar] [CrossRef]
  17. E Walker, R.; Bechtel, C.M.; Natarajan, V.; Baseler, M.; Hege, K.M.; A Metcalf, J.; Stevens, R.; Hazen, A.; Blaese, R.M.; Chen, C.C.; et al. Long-term in vivo survival of receptor-modified syngeneic T cells in patients with human immunodeficiency virus infection. Blood 2000, 96, 467–474. [Google Scholar]
  18. Mitsuyasu, R.T.; A Anton, P.; Deeks, S.G.; Scadden, D.T.; Connick, E.; Downs, M.T.; Bakker, A.; Roberts, M.R.; June, C.H.; Jalali, S.; et al. Prolonged survival and tissue trafficking following adoptive transfer of CD4zeta gene-modified autologous CD4(+) and CD8(+) T cells in human immunodeficiency virus-infected subjects. Blood 2000, 96, 785–793. [Google Scholar] [CrossRef]
  19. Hudecek, M.; Sommermeyer, D.; Kosasih, P.L.; Silva-Benedict, A.; Liu, L.; Rader, C.; Jensen, M.C.; Riddell, S.R. The Nonsignaling Extracellular Spacer Domain of Chimeric Antigen Receptors Is Decisive for In Vivo Antitumor Activity. Cancer Immunol. Res. 2014, 3, 125–135. [Google Scholar] [CrossRef] [Green Version]
  20. Berger, C.; Jensen, M.C.; Lansdorp, P.M.; Gough, M.; Elliott, C.; Riddell, S.R. Adoptive transfer of effector CD8+ T cells derived from central memory cells establishes persistent T cell memory in primates. J. Clin. Investig. 2008, 118, 294–305. [Google Scholar] [CrossRef] [Green Version]
  21. Berger, C.; Berger, M.; Anderson, D.; Riddell, S.R. A non-human primate model for analysis of safety, persistence, and function of adoptively transferred T cells. J. Med Primatol. 2010, 40, 88–103. [Google Scholar] [CrossRef] [PubMed]
  22. Klebanoff, C.A.; Gattinoni, L.; Torabi-Parizi, P.; Kerstann, K.; Cardones, A.R.; Finkelstein, S.E.; Palmer, D.C.; Antony, P.A.; Hwang, S.T.; Rosenberg, S.A.; et al. Central memory self/tumor-reactive CD8+ T cells confer superior antitumor immunity compared with effector memory T cells. Proc. Natl. Acad. Sci. USA 2005, 102, 9571–9576. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  23. Hinrichs, C.S.; Borman, Z.A.; Cassard, L.; Gattinoni, L.; Spolski, R.; Yu, Z.; Sanchez-Perez, L.; Muranski, P.; Kern, S.J.; Logun, C.; et al. Adoptively transferred effector cells derived from naïve rather than central memory CD8+ T cells mediate superior antitumor immunity. Proc. Natl. Acad. Sci. USA 2009, 106, 17469–17474. [Google Scholar] [CrossRef] [Green Version]
  24. Westwood, J.A.; Smyth, M.J.; Teng, M.W.; Moeller, M.; Trapani, J.A.; Scott, A.M.; Smyth, F.E.; Cartwright, G.A.; Power, B.; Hönemann, D.; et al. Adoptive transfer of T cells modified with a humanized chimeric receptor gene inhibits growth of Lewis-Y-expressing tumors in mice. Proc. Natl. Acad. Sci. USA 2005, 102, 19051–19056. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  25. Moeller, M.; Kershaw, M.; Cameron, R.; A Westwood, J.; Trapani, J.A.; Smyth, M.J.; Darcy, P.K. Sustained Antigen-Specific Antitumor Recall Response Mediated by Gene-Modified CD4+ T Helper-1 and CD8+ T Cells. Cancer Res. 2007, 67, 11428–11437. [Google Scholar] [CrossRef] [Green Version]
  26. Turtle, C.J.; Hanafi, L.-A.; Berger, C.; Gooley, T.A.; Cherian, S.; Hudecek, M.; Sommermeyer, D.; Melville, K.; Pender, B.; Budiarto, T.M.; et al. CD19 CAR-T cells of defined CD4+:CD8+ composition in adult B cell ALL patients. J. Clin. Investig. 2016, 126, 2123–2138. [Google Scholar] [CrossRef] [Green Version]
  27. Lim, W.A.; June, C.H. The Principles of Engineering Immune Cells to Treat Cancer. Cell 2017, 168, 724–740. [Google Scholar] [CrossRef] [Green Version]
  28. Danilova, L.V.; Anagnostou, V.; Caushi, J.X.; Sidhom, J.-W.; Guo, H.; Chan, H.Y.; Suri, P.; Tam, A.J.; Zhang, J.; El Asmar, M.; et al. The Mutation-Associated Neoantigen Functional Expansion of Specific T Cells (MANAFEST) Assay: A Sensitive Platform for Monitoring Antitumor Immunity. Cancer Immunol. Res. 2018. [Google Scholar] [CrossRef] [Green Version]
  29. Brudno, J.N.; Kochenderfer, J.N. Chimeric antigen receptor T-cell therapies for lymphoma. Nat. Rev. Clin. Oncol. 2017, 15, 31–46. [Google Scholar] [CrossRef]
  30. Smith, M.; Zakrzewski, J.; James, S.; Sadelain, M. Posttransplant chimeric antigen receptor therapy. Blood 2018, 131, 1045–1052. [Google Scholar] [CrossRef] [Green Version]
  31. Dotti, G.; Gottschalk, S.; Savoldo, B.; Brenner, M.K. Design and development of therapies using chimeric antigen receptor-expressing T cells. Immunol. Rev. 2014, 257, 107–126. [Google Scholar] [CrossRef] [PubMed]
  32. Watanabe, K.; Kuramitsu, S.; Posey, A.D.; June, C.H. Expanding the Therapeutic Window for CAR T Cell Therapy in Solid Tumors: The Knowns and Unknowns of CAR T Cell Biology. Front. Immunol. 2018, 9, 2486. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  33. D’Aloia, M.M.; Zizzari, I.G.; Sacchetti, B.; Pierelli, L.; Alimandi, M. CAR-T cells: The long and winding road to solid tumors. Cell Death Dis. 2018, 9, 282. [Google Scholar] [CrossRef]
  34. Lamers, C.H.J.; Sleijfer, S.; Van Steenbergen, S.; Van Elzakker, P.; Van Krimpen, B.; Groot, C.; Vulto, A.; Bakker, M.D.; Oosterwijk, E.; Debets, R.; et al. Treatment of Metastatic Renal Cell Carcinoma With CAIX CAR-engineered T cells: Clinical Evaluation and Management of On-target Toxicity. Mol. Ther. 2013, 21, 904–912. [Google Scholar] [CrossRef]
  35. Perales, M.A.; Kebriaei, P.; Kean, L.S.; Sadelain, M. Building a Safer and Faster CAR: Seatbelts, Airbags, and CRISPR. Boil. Blood Marrow Transplant. 2017, 24, 27–31. [Google Scholar] [CrossRef] [Green Version]
  36. Harrer, D.C.; Dörrie, J.; Schaft, N. Chimeric Antigen Receptors in Different Cell Types: New Vehicles Join the Race. Hum. Gene Ther. 2018, 29, 547–558. [Google Scholar] [CrossRef]
  37. Vantourout, P.; Hayday, A. Six-of-the-best: Unique contributions of gammadelta T cells to immunology. Nat. Rev. Immunol. 2013, 13, 88–100. [Google Scholar] [CrossRef] [Green Version]
  38. Morita, C.; Mariuzza, R.A.; Brenner, M.B. Antigen recognition by human γδ T cells: Pattern recognition by the adaptive immune system. Springer Semin. Immunopathol. 2000, 22, 191–217. [Google Scholar] [CrossRef]
  39. Holtmeier, W.; Kabelitz, D. γδ T Cells Link Innate and Adaptive Immune Responses. Chem. Immunol. Allergy 2005, 86, 151–183. [Google Scholar] [CrossRef]
  40. Hilden, J.M.; Dinndorf, P.A.; Meerbaum, S.O.; Sather, H.; Villaluna, D.; Heerema, N.A.; McGlennen, R.; Smith, F.O.; Woods, W.G.; Salzer, W.L.; et al. Analysis of prognostic factors of acute lymphoblastic leukemia in infants: Report on CCG 1953 from the Children’s Oncology Group. Blood 2006, 108, 441–451. [Google Scholar] [CrossRef]
  41. Mirzaei, H.R.; Mirzaei, H.; Lee, S.Y.; Hadjati, J.; Till, B. Prospects for chimeric antigen receptor (CAR) γδ T cells: A potential game changer for adoptive T cell cancer immunotherapy. Cancer Lett. 2016, 380, 413–423. [Google Scholar] [CrossRef] [Green Version]
  42. Zhao, Y.; Niu, C.; Cui, J. Gamma-delta (γδ) T cells: Friend or foe in cancer development? J. Transl. Med. 2018, 16, 3. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  43. Silva-Santos, B. Promoting angiogenesis within the tumor microenvironment: The secret life of murine lymphoid IL-17-producing γδ T cells. Eur. J. Immunol. 2010, 40, 1873–1876. [Google Scholar] [CrossRef] [PubMed]
  44. Paul, S.; Lal, G. Regulatory and effector functions of gamma-delta (γδ) T cells and their therapeutic potential in adoptive cellular therapy for cancer. Int. J. Cancer 2016, 139, 976–985. [Google Scholar] [CrossRef] [PubMed]
  45. Peng, W.; Chen, J.Q.; Liu, C.; Malu, S.; Creasy, C.; Tetzlaff, M.T.; Xu, C.; McKenzie, J.A.; Zhang, C.; Liang, X.; et al. Loss of PTEN Promotes Resistance to T Cell-Mediated Immunotherapy. Cancer Discov. 2015, 6, 202–216. [Google Scholar] [CrossRef] [Green Version]
  46. Qu, P.; Wang, L.-Z.; Lin, P.C. Expansion and functions of myeloid-derived suppressor cells in the tumor microenvironment. Cancer Lett. 2016, 380, 253–256. [Google Scholar] [CrossRef]
  47. Oevermann, L.; Lang, P.; Feuchtinger, T.; Schumm, M.; Teltschik, H.-M.; Schlegel, P.; Handgretinger, R. Immune reconstitution and strategies for rebuilding the immune system after haploidentical stem cell transplantation. Ann. New York Acad. Sci. 2012, 1266, 161–170. [Google Scholar] [CrossRef]
  48. Critchley-Thorne, R.J.; Simons, D.L.; Yan, N.; Miyahira, A.K.; Dirbas, F.M.; Johnson, D.L.; Swetter, S.M.; Carlson, R.W.; Fisher, G.A.; Koong, A.; et al. Impaired interferon signaling is a common immune defect in human cancer. Proc. Natl. Acad. Sci. USA 2009, 106, 9010–9015. [Google Scholar] [CrossRef] [Green Version]
  49. Brandes, M. Professional Antigen-Presentation Function by Human gd T Cells. Science 2005, 309, 264–268. [Google Scholar] [CrossRef]
  50. Muto, M.; Baghdadi, M.; Maekawa, R.; Wada, H.; Seino, K.-I. Myeloid molecular characteristics of human γδ T cells support their acquisition of tumor antigen-presenting capacity. Cancer Immunol. Immunother. 2015, 64, 941–949. [Google Scholar] [CrossRef] [Green Version]
  51. Godfrey, D.; Stankovic, S.; Baxter, A. Raising the NKT cell family. Nat. Immunol. 2010, 11, 197–206. [Google Scholar] [CrossRef]
  52. Godfrey, D.; Macdonald, H.R.; Kronenberg, M.; Smyth, M.J.; Van Kaer, L. NKT cells: What’s in a name? Nat. Rev. Immunol. 2004, 4, 231–237. [Google Scholar] [CrossRef] [PubMed]
  53. Gumperz, J.E.; Miyake, S.; Yamamura, T.; Brenner, M.B. Functionally Distinct Subsets of CD1d-restricted Natural Killer T Cells Revealed by CD1d Tetramer Staining. J. Exp. Med. 2002, 195, 625–636. [Google Scholar] [CrossRef]
  54. Tachibana, T.; Onodera, H.; Tsuruyama, T.; Imamura, M.; Mori, A.; Nagayama, S.; Hiai, H. Increased Intratumor V?24-Positive Natural Killer T Cells: A Prognostic Factor for Primary Colorectal Carcinomas. Clin. Cancer Res. 2005, 11, 7322–7327. [Google Scholar] [CrossRef] [Green Version]
  55. Terabe, M.; Matsui, S.; Noben-Trauth, N.; Chen, H.; Watson, C.; Donaldson, D.D.; Carbone, D.P.; Paul, W.E.; Berzofsky, J.A. NKT cell–mediated repression of tumor immunosurveillance by IL-13 and the IL-4R–STAT6 pathway. Nat. Immunol. 2000, 1, 515–520. [Google Scholar] [CrossRef]
  56. Yu, J. Antitumor Activity of T Cells Generated from Lymph Nodes Draining the SEA-expressing Murine B16 Melanoma and Secondarily Activated with Dendritic Cells. Int. J. Boil. Sci. 2009, 5, 135–146. [Google Scholar] [CrossRef] [PubMed]
  57. Gottschalk, C.; Mettke, E.; Kurts, C. The Role of Invariant Natural Killer T Cells in Dendritic Cell Licensing, Cross-Priming, and Memory CD8+ T Cell Generation. Front. Immunol. 2015, 6, 379. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  58. Morvan, M.G.; Lanier, L.L. NK cells and cancer: You can teach innate cells new tricks. Nat. Rev. Cancer 2015, 16, 7–19. [Google Scholar] [CrossRef]
  59. Long, G.V.; Stroyakovskiy, D.; Gogas, H.; Levchenko, E.V.; De Braud, F.; Larkin, J.; Garbe, C.; Jouary, T.; Hauschild, A.; Grob, J.-J.; et al. Dabrafenib and trametinib versus dabrafenib and placebo for Val600 BRAF-mutant melanoma: A multicentre, double-blind, phase 3 randomised controlled trial. Lancet 2015, 386, 444–451. [Google Scholar] [CrossRef]
  60. Bollino, D.; Webb, T.J. Chimeric antigen receptor-engineered natural killer and natural killer T cells for cancer immunotherapy. Transl. Res. 2017, 187, 32–43. [Google Scholar] [CrossRef]
  61. Ruggeri, L.; Capanni, M.; Urbani, E.; Shlomchik, W.D.; Posati, S.; Rogaia, D.; Martelli, M.F.; Velardi, A.; Perruccio, K.; Tosti, A.; et al. Effectiveness of Donor Natural Killer Cell Alloreactivity in Mismatched Hematopoietic Transplants. Science 2002, 295, 2097–2100. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  62. Miller, J.S.; Soignier, Y.; Panoskaltsis-Mortari, A.; McNearney, S.A.; Yun, G.H.; Fautsch, S.K.; McKenna, D.; Le, C.; DeFor, T.; Burns, L.J.; et al. Successful adoptive transfer and in vivo expansion of human haploidentical NK cells in patients with cancer. Blood 2005, 105, 3051–3057. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  63. Klöß, S.; Oberschmidt, O.; A Morgan, M.; Dahlke, J.; Arseniev, L.; Huppert, V.; Granzin, M.; Gardlowski, T.; Matthies, N.; Soltenborn, S.; et al. Optimization of Human NK Cell Manufacturing: Fully Automated Separation, ImprovedEx VivoExpansion Using IL-21 with Autologous Feeder Cells, and Generation of Anti-CD123-CAR-Expressing Effector Cells. Hum. Gene Ther. 2017, 28, 897–913. [Google Scholar] [CrossRef]
  64. Bosch-Voskens, C.; Watanabe, R.; Rollins, S.; Campana, D.; Hasumi, K.; Mann, D. Ex-vivo expanded human NK cells express activating receptors that mediate cytotoxicity of allogeneic and autologous cancer cell lines by direct recognition and antibody directed cellular cytotoxicity. J. Exp. Clin. Cancer Res. 2010, 29, 134. [Google Scholar] [CrossRef] [Green Version]
  65. Sutlu, T.; Stellan, B.; Gilljam, M.; Quezada, H.C.; Nahi, H.H.; Gahrton, G.; Alici, E. Clinical-grade, large-scale, feeder-free expansion of highly active human natural killer cells for adoptive immunotherapy using an automated bioreactor. Cytotherapy 2010, 12, 1044–1055. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  66. Liu, E.; Tong, Y.; Dotti, G.; Shaim, H.; Savoldo, B.; Mukherjee, M.; Orange, J.; Wan, X.; Lu, X.; Reynolds, A.; et al. Cord blood NK cells engineered to express IL-15 and a CD19-targeted CAR show long-term persistence and potent antitumor activity. Leukemia 2017, 32, 520–531. [Google Scholar] [CrossRef]
  67. Lowe, E.; Truscott, L.C.; De Oliveira, S. In Vitro Generation of Human NK Cells Expressing Chimeric Antigen Receptor Through Differentiation of Gene-Modified Hematopoietic Stem Cells. Breast Cancer 2016, 1441, 241–251. [Google Scholar] [CrossRef] [Green Version]
  68. Li, Y.; Hermanson, D.L.; Moriarity, B.S.; Kaufman, D. Human iPSC-Derived Natural Killer Cells Engineered with Chimeric Antigen Receptors Enhance Anti-Tumor Activity. Cell Stem Cell 2018, 23, 181–192. [Google Scholar] [CrossRef] [Green Version]
  69. Oelsner, S.; Friede, M.E.; Zhang, C.; Wagner, J.; Badura, S.; Bader, P.; Ullrich, E.; Ottmann, O.; Klingemann, H.; Tonn, T.; et al. Continuously expanding CAR NK-92 cells display selective cytotoxicity against B-cell leukemia and lymphoma. Cytotherapy 2017, 19, 235–249. [Google Scholar] [CrossRef] [Green Version]
  70. Klingemann, H.; Boissel, L.; Toneguzzo, F. Natural Killer Cells for Immunotherapy—Advantages of the NK-92 Cell Line over Blood NK Cells. Front. Immunol. 2016, 7, 177. [Google Scholar] [CrossRef] [Green Version]
  71. Yong, C.S.; John, L.B.; Devaud, C.; Prince, M.H.; Johnstone, R.W.; Trapani, J.A.; Darcy, P.K.; Kershaw, M. A role for multiple chimeric antigen receptor-expressing leukocytes in antigen-specific responses to cancer. Oncotarget 2016, 7, 34582–34598. [Google Scholar] [CrossRef] [PubMed]
  72. Hu, Y.; Tian, Z.-G.; Zhang, C. Chimeric antigen receptor (CAR)-transduced natural killer cells in tumor immunotherapy. Acta Pharmacol. Sin. 2017, 39, 167–176. [Google Scholar] [CrossRef]
  73. Ramos, C.A.; Rouce, R.; Robertson, C.S.; Reyna, A.; Narala, N.; Vyas, G.; Mehta, B.; Zhang, H.; Dakhova, O.; Carrum, G.; et al. In Vivo Fate and Activity of Second- versus Third-Generation CD19-Specific CAR-T Cells in B Cell Non-Hodgkin’s Lymphomas. Mol. Ther. 2018, 26, 2727–2737. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  74. Liu, E.; Marin, D.; Banerjee, P.; Macapinlac, H.A.; Thompson, P.; Basar, R.; Kerbauy, L.N.; Overman, B.; Thall, P.; Kaplan, M.; et al. Use of CAR-Transduced Natural Killer Cells in CD19-Positive Lymphoid Tumors. N. Engl. J. Med. 2020, 382, 545–553. [Google Scholar] [CrossRef] [PubMed]
  75. De Oliveira, S.; Ryan, C.; Giannoni, F.; Hardee, C.L.; Tremcinska, I.; Katebian, B.; Wherley, J.; Sahaghian, A.; Tu, A.; Grogan, T.; et al. Modification of Hematopoietic Stem/Progenitor Cells with CD19-Specific Chimeric Antigen Receptors as a Novel Approach for Cancer Immunotherapy. Hum. Gene Ther. 2013, 24, 824–839. [Google Scholar] [CrossRef] [PubMed]
  76. Larson, S.; De Oliveira, S. Gene-modified hematopoietic stem cells for cancer immunotherapy. Hum. Vaccines Immunother. 2014, 10, 982–985. [Google Scholar] [CrossRef] [Green Version]
  77. Gschweng, E.; De Oliveira, S.; Kohn, D.B. Hematopoietic stem cells for cancer immunotherapy. Immunol. Rev. 2014, 257, 237–249. [Google Scholar] [CrossRef] [Green Version]
  78. Larson, S.M.; Truscott, L.C.; Chiou, T.-T.; Patel, A.; Kao, R.; Tu, A.; Tyagi, T.; Lu, X.; Elashoff, D.; De Oliveira, S. Pre-clinical development of gene modification of haematopoietic stem cells with chimeric antigen receptors for cancer immunotherapy. Hum. Vaccines Immunother. 2017, 13, 1094–1104. [Google Scholar] [CrossRef]
  79. Kao, R.L.; Truscott, L.C.; Chiou, T.-T.; Tsai, W.; Wu, A.M.; De Oliveira, S. A Cetuximab-Mediated Suicide System in Chimeric Antigen Receptor-Modified Hematopoietic Stem Cells for Cancer Therapy. Hum. Gene Ther. 2019, 30, 413–428. [Google Scholar] [CrossRef]
  80. Hege, K.M.; Cooke, K.S.; Finer, M.H.; Zsebo, K.M.; Roberts, M.R. Systemic T Cell–independent Tumor Immunity after Transplantation of Universal Receptor–modified Bone Marrow into SCID Mice. J. Exp. Med. 1996, 184, 2261–2270. [Google Scholar] [CrossRef] [Green Version]
  81. Roberts, M.R.; Cooke, K.S.; Tran, A.C.; A Smith, K.; Lin, W.Y.; Wang, M.; Dull, T.J.; Farson, D.; Zsebo, K.M.; Finer, M.H. Antigen-specific cytolysis by neutrophils and NK cells expressing chimeric immune receptors bearing zeta or gamma signaling domains. J. Immunol. 1998, 161, 375–384. [Google Scholar] [PubMed]
  82. Lin, W.Y.; Roberts, M.R. Developmental dissociation of T cells from B, NK, and myeloid cells revealed by MHC class II–specific chimeric immune receptors bearing TCR-ζ or FcR-γ chain signaling domains. Blood 2002, 100, 3045–3048. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  83. Witko-Sarsat, V.; Rieu, P.; Descamps-Latscha, B.; Lesavre, P.; Halbwachs-Mecarelli, L. Neutrophils: Molecules, functions and pathophysiological aspects. Lab. Investig. 2000, 80, 617–653. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  84. Nichols, B.A.; Bainton, D.F.; Farquhar, M.G. Differentiation of Monocytes. J. Cell Boil. 1971, 50, 498–515. [Google Scholar] [CrossRef] [PubMed]
  85. Biglari, A.; Southgate, T.D.; Fairbairn, L.J.; E Gilham, D. Human monocytes expressing a CEA-specific chimeric CD64 receptor specifically target CEA-expressing tumour cells in vitro and in vivo. Gene Ther. 2006, 13, 602–610. [Google Scholar] [CrossRef] [PubMed]
  86. Wang, L.-X.; Zhang, S.-X.; Wu, H.-J.; Rong, X.-L.; Guo, J. M2b macrophage polarization and its roles in diseases. J. Leukoc. Boil. 2018, 106, 345–358. [Google Scholar] [CrossRef] [Green Version]
  87. Mosser, D.; Edwards, J. Exploring the full spectrum of macrophage activation. Nat. Rev. Immunol. 2008, 8, 958–969. [Google Scholar] [CrossRef]
  88. Velmurugan, R.; Challa, D.K.; Ram, S.; Ober, R.; Ward, E.S. Macrophage-Mediated Trogocytosis Leads to Death of Antibody-Opsonized Tumor Cells. Mol. Cancer Ther. 2016, 15, 1879–1889. [Google Scholar] [CrossRef] [Green Version]
  89. Klichinsky, M.; Ruella, M.; Shestova, O.; Kenderian, S.S.; Kim, M.Y.; O’Connor, R.; Scholler, J.; June, C.; Gill, S. Abstract 4575: Chimeric antigen receptor macrophages (CARMA) for adoptive cellular immunotherapy of solid tumors. Immunology 2017, 77, 4575. [Google Scholar] [CrossRef]
  90. Morrissey, M.A.; Williamson, A.P.; Steinbach, A.; Roberts, E.; Kern, N.; Headley, M.B.; Vale, R.D. Chimeric antigen receptors that trigger phagocytosis. eLife 2018, 7, e36688. [Google Scholar] [CrossRef]
  91. Chanmee, T.; Ontong, P.; Konno, K.; Itano, N. Tumor-Associated Macrophages as Major Players in the Tumor Microenvironment. Cancers 2014, 6, 1670–1690. [Google Scholar] [CrossRef] [Green Version]
  92. Smith, E.W.C.; Weagel, E.; Smith, C.; Liu, P.G.; Robison, R.; O’Neill, K. Macrophage Polarization and Its Role in Cancer. J. Clin. Cell. Immunol. 2015, 6, 1–8. [Google Scholar] [CrossRef]
  93. Bingle, L.; Brown, N.J.; Lewis, C. The role of tumour-associated macrophages in tumour progression: Implications for new anticancer therapies. J. Pathol. 2002, 196, 254–265. [Google Scholar] [CrossRef]
  94. Murdoch, C.; Giannoudis, A.; Lewis, C.E. Mechanisms regulating the recruitment of macrophages into hypoxic areas of tumors and other ischemic tissues. Blood 2004, 104, 2224–2234. [Google Scholar] [CrossRef]
  95. Valipour, B.; Velaei, K.; Abedelahi, A.; Karimipour, M.; Darabi, M.; Charoudeh, H.N. NK cells: An attractive candidate for cancer therapy. J. Cell. Physiol. 2019, 234, 19352–19365. [Google Scholar] [CrossRef] [PubMed]
  96. Tseng, D.; Volkmer, J.-P.; Willingham, S.B.; Contreras-Trujillo, H.; Fathman, J.W.; Fernhoff, N.B.; Seita, J.; Inlay, M.A.; Weiskopf, K.; Miyanishi, M.; et al. Anti-CD47 antibody-mediated phagocytosis of cancer by macrophages primes an effective antitumor T-cell response. Proc. Natl. Acad. Sci. USA 2013, 110, 11103–11108. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  97. Asano, K.; Nabeyama, A.; Miyake, Y.; Qiu, C.-H.; Kurita, A.; Tomura, M.; Kanagawa, O.; Fujii, S.-I.; Tanaka, M. CD169-Positive Macrophages Dominate Antitumor Immunity by Crosspresenting Dead Cell-Associated Antigens. Immunity1 2011, 34, 85–95. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  98. Velazquez, E.J.; Lattin, J.E.; Brindley, T.D.; Reinstein, Z.Z.; Chu, R.; Liu, L.; Weagel, E.G.; Townsend, M.H.; Whitley, K.V.; Lawrence, E.L.; et al. Abstract 2563: Macrophage Toll-like receptor-chimeric antigen receptors (MOTO-CARs) as a novel adoptive cell therapy for the treatment of solid malignancies. Immunology 2018, 78, 2563. [Google Scholar] [CrossRef]
  99. Najafi, M.; Goradel, N.H.; Farhood, B.; Salehi, E.; Nashtaei, M.S.; Khanlarkhani, N.; Khezri, Z.; Majidpoor, J.; Mortezaee, K.; Habibi, M.; et al. Macrophage polarity in cancer: A review. J. Cell. Biochem. 2018, 120, 2756–2765. [Google Scholar] [CrossRef] [PubMed]
  100. Zhang, X.; Edwards, J.; Mosser, D.M. The Expression of Exogenous Genes in Macrophages: Obstacles and Opportunities. Breast Cancer 2009, 531, 123–143. [Google Scholar] [CrossRef] [Green Version]
  101. Schuler, G.; Schuler-Thurner, B.; Steinman, R.M. The use of dendritic cells in cancer immunotherapy. Curr. Opin. Immunol. 2003, 15, 138–147. [Google Scholar] [CrossRef]
  102. Wu, M.; Zhang, L.; Zhang, H.; Ning, J.; Tu, S.; He, Y.; Li, Y. CD19 chimeric antigen receptor–redirected T cells combined with epidermal growth factor receptor pathway substrate 8 peptide–derived dendritic cell vaccine in leukemia. Cytotherapy 2019, 21, 659–670. [Google Scholar] [CrossRef] [PubMed]
  103. Squadrito, M.L.; Cianciaruso, C.; Hansen, S.K.; De Palma, M. EVIR: Chimeric receptors that enhance dendritic cell cross-dressing with tumor antigens. Nat. Methods 2018, 15, 183–186. [Google Scholar] [CrossRef] [PubMed]
  104. Eshhar, Z.; Waks, T.; Gross, G.; Schindler, D.G. Specific activation and targeting of cytotoxic lymphocytes through chimeric single chains consisting of antibody-binding domains and the gamma or zeta subunits of the immunoglobulin and T-cell receptors. Proc. Natl. Acad. Sci. USA 1993, 90, 720–724. [Google Scholar] [CrossRef] [Green Version]
  105. Savoldo, B.; Ramos, C.A.; Liu, E.; Mims, M.P.; Keating, M.J.; Carrum, G.; Kamble, R.T.; Bollard, C.M.; Gee, A.P.; Mei, Z.; et al. CD28 costimulation improves expansion and persistence of chimeric antigen receptor-modified T cells in lymphoma patients. J. Clin. Investig. 2011, 121, 1822–1826. [Google Scholar] [CrossRef] [Green Version]
  106. Long, A.H.; Haso, W.M.; Shern, J.F.; Wanhainen, K.; Murgai, M.; Ingaramo, M.; Smith, J.P.; Walker, A.J.; Kohler, M.E.; Venkateshwara, V.R.; et al. 4-1BB costimulation ameliorates T cell exhaustion induced by tonic signaling of chimeric antigen receptors. Nat. Med. 2015, 21, 581–590. [Google Scholar] [CrossRef] [Green Version]
  107. Zhao, Z.; Condomines, M.; Van Der Stegen, S.J.; Perna, F.; Kloss, C.C.; Gunset, G.; Plotkin, J.; Sadelain, M. Structural Design of Engineered Costimulation Determines Tumor Rejection Kinetics and Persistence of CAR T Cells. Cancer Cell 2015, 28, 415–428. [Google Scholar] [CrossRef] [Green Version]
  108. Zabel, M.; Tauber, P.A.; Pickl, W.F. The making and function of CAR cells. Immunol. Lett. 2019, 212, 53–69. [Google Scholar] [CrossRef]
  109. Loskog, A.; Giandomenico, V.; Rossig, C.; Pule, M.; Dotti, G.; Brenner, M.K. Addition of the CD28 signaling domain to chimeric T-cell receptors enhances chimeric T-cell resistance to T regulatory cells. Leukemia 2006, 20, 1819–1828. [Google Scholar] [CrossRef]
  110. Song, D.-G.; Ye, Q.; Poussin, M.; Harms, G.M.; Figini, M.; Powell, D.J. CD27 costimulation augments the survival and antitumor activity of redirected human T cells in vivo. Blood 2012, 119, 696–706. [Google Scholar] [CrossRef]
  111. Cheadle, E.J.; Sheard, V.; Hombach, A.A.; Chmielewski, M.; Riet, T.; Berrevoets, C.; Schooten, E.; Lamers, C.; Abken, H.; Debets, R.; et al. Chimeric Antigen Receptors for T-Cell Based Therapy. Adv. Struct. Saf. Stud. 2012, 907, 645–666. [Google Scholar] [CrossRef]
  112. Guedan, S.; Chen, X.; Madar, A.; Carpenito, C.; Mcgettigan, S.E.; Frigault, M.J.; Lee, J.; Posey, A.D.; Scholler, J.; Scholler, N.; et al. ICOS-based chimeric antigen receptors program bipolar TH17/TH1 cells. Blood 2014, 124, 1070–1080. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  113. Kawalekar, O.U.; O’Connor, R.S.; Fraietta, J.A.; Guo, L.; Mcgettigan, S.E.; Posey, A.D.; Patel, P.R.; Guedan, S.; Scholler, J.; Keith, B.; et al. Distinct Signaling of Coreceptors Regulates Specific Metabolism Pathways and Impacts Memory Development in CAR T Cells. Immunity 2016, 44, 380–390. [Google Scholar] [CrossRef] [PubMed]
  114. Gomes-Silva, D.; Mukherjee, M.; Srinivasan, M.; Krenciute, G.; Dakhova, O.; Zheng, Y.; Cabral, J.M.S.; Rooney, C.M.; Orange, J.S.; Brenner, M.K.; et al. Tonic 4-1BB Costimulation in Chimeric Antigen Receptors Impedes T Cell Survival and Is Vector-Dependent. Cell Rep. 2017, 21, 17–26. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  115. Li, G.; Boucher, J.C.; Kotani, H.; Park, K.; Zhang, Y.; Shrestha, B.; Wang, X.; Guan, L.; Beatty, N.; Abate-Daga, D.; et al. 4-1BB enhancement of CAR T function requires NF-κB and TRAFs. JCI Insight 2018, 3. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  116. Mamonkin, M.; Mukherjee, M.; Srinivasan, M.; Sharma, S.; Gomes-Silva, D.; Mo, F.; Krenciute, G.; Orange, J.S.; Brenner, M.K. Reversible Transgene Expression Reduces Fratricide and Permits 4-1BB Costimulation of CAR T Cells Directed to T-cell Malignancies. Cancer Immunol. Res. 2017, 6, 47–58. [Google Scholar] [CrossRef] [Green Version]
  117. Salter, A.I.; Ivey, R.G.; Kennedy, J.J.; Voillet, V.; Rajan, A.; Alderman, E.J.; Voytovich, U.J.; Lin, C.; Sommermeyer, D.; Liu, L.; et al. Phosphoproteomic analysis of chimeric antigen receptor signaling reveals kinetic and quantitative differences that affect cell function. Sci. Signal. 2018, 11, eaat6753. [Google Scholar] [CrossRef] [Green Version]
  118. Golumba-Nagy, V.; Kuehle, J.; Hombach, A.A.; Abken, H. CD28-ζ CAR T Cells Resist TGF-β Repression through IL-2 Signaling, Which Can Be Mimicked by an Engineered IL-7 Autocrine Loop. Mol. Ther. 2018, 26, 2218–2230. [Google Scholar] [CrossRef] [Green Version]
  119. Priceman, S.J.; Gerdts, E.A.; Tilakawardane, D.; Kennewick, K.T.; Murad, J.; Park, A.K.; Jeang, B.; Yamaguchi, Y.; Yang, X.; Urak, R.; et al. Co-stimulatory signaling determines tumor antigen sensitivity and persistence of CAR T cells targeting PSCA+ metastatic prostate cancer. OncoImmunology 2017, 7, e1380764. [Google Scholar] [CrossRef]
  120. Ramello, M.C.; Benzaïd, I.; Kuenzi, B.M.; Lienlaf-Moreno, M.; Kandell, W.M.; Santiago, D.N.; Pabón-Saldaña, M.; Darville, L.; Fang, B.; Rix, U.; et al. An immunoproteomic approach to characterize the CAR interactome and signalosome. Sci. Signal. 2019, 12, eaap9777. [Google Scholar] [CrossRef]
  121. Roselli, E.; Frieling, J.S.; Thorner, K.; Ramello, M.C.; Lynch, C.C.; Abate-Daga, D. CAR-T Engineering: Optimizing Signal Transduction and Effector Mechanisms. BioDrugs 2019, 33, 647–659. [Google Scholar] [CrossRef] [PubMed]
  122. Guedan, S.; Posey, A.D.; Shaw, C.; Wing, A.; Da, T.; Patel, P.R.; Mcgettigan, S.E.; Casado-Medrano, V.; Kawalekar, O.U.; Uribe-Herranz, M.; et al. Enhancing CAR T cell persistence through ICOS and 4-1BB costimulation. JCI Insight 2018, 3. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  123. Beard, R.E.; Zheng, Z.; Lagisetty, K.H.; Burns, W.R.; Tran, E.; Hewitt, S.M.; Abate-Daga, D.; Rosati, S.F.; Fine, H.A.; Ferrone, S.; et al. Multiple chimeric antigen receptors successfully target chondroitin sulfate proteoglycan 4 in several different cancer histologies and cancer stem cells. J. Immunother. Cancer 2014, 2, 25. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  124. Zhong, X.-S.; Matsushita, M.; Plotkin, J.; Rivière, I.; Sadelain, M. Chimeric Antigen Receptors Combining 4-1BB and CD28 Signaling Domains Augment PI3kinase/AKT/Bcl-XL Activation and CD8+ T Cell–mediated Tumor Eradication. Mol. Ther. 2009, 18, 413–420. [Google Scholar] [CrossRef]
  125. Till, B.G.; Jensen, M.C.; Wang, J.; Qian, X.; Gopal, A.K.; Maloney, D.G.; Lindgren, C.G.; Lin, Y.; Pagel, J.M.; Budde, L.E.; et al. CD20-specific adoptive immunotherapy for lymphoma using a chimeric antigen receptor with both CD28 and 4-1BB domains: Pilot clinical trial results. Blood 2012, 119, 3940–3950. [Google Scholar] [CrossRef] [Green Version]
  126. Lesch, S.; Benmebarek, M.-R.; Cadilha, B.L.; Stoiber, S.; Subklewe, M.; Endres, S.; Kobold, S. Determinants of response and resistance to CAR T cell therapy. Semin. Cancer Boil. 2019. [Google Scholar] [CrossRef]
  127. Guest, R.D.; E Hawkins, R.; Kirillova, N.; Cheadle, E.J.; Arnold, J.; O??neill, A.; Irlam, J.; A Chester, K.; Kemshead, J.T.; Shaw, D.M.; et al. The Role of Extracellular Spacer Regions in the Optimal Design of Chimeric Immune Receptors. J. Immunother. 2005, 28, 203–211. [Google Scholar] [CrossRef]
  128. Watanabe, N.; Bajgain, P.; Sukumaran, S.; Ansari, S.; Heslop, H.E.; Rooney, C.M.; Brenner, M.K.; Leen, A.M.; Vera, J.F. Fine-tuning the CAR spacer improves T-cell potency. OncoImmunology 2016, 5, e1253656. [Google Scholar] [CrossRef] [Green Version]
  129. Alabanza, L.; Pegues, M.; Geldres, C.; Shi, V.; Wiltzius, J.J.; Sievers, S.A.; Yang, S.; Kochenderfer, J.N. Function of Novel Anti-CD19 Chimeric Antigen Receptors with Human Variable Regions Is Affected by Hinge and Transmembrane Domains. Mol. Ther. 2017, 25, 2452–2465. [Google Scholar] [CrossRef] [Green Version]
  130. Feucht, J.; Sun, J.; Eyquem, J.; Ho, Y.-J.; Zhao, Z.; Leibold, J.; Dobrin, A.; Cabriolu, A.; Hamieh, M.; Sadelain, M. Calibration of CAR activation potential directs alternative T cell fates and therapeutic potency. Nat. Med. 2018, 25, 82–88. [Google Scholar] [CrossRef]
  131. Call, M.E.; Pyrdol, J.; Wiedmann, M.; Wucherpfennig, K.W. The Organizing Principle in the Formation of the T Cell Receptor-CD3 Complex. Cell 2002, 111, 967–979. [Google Scholar] [CrossRef] [Green Version]
  132. Dong, D.; Zheng, L.; Lin, J.; Zhang, B.; Zhu, Y.; Li, N.; Xie, S.; Wang, Y.; Gao, N.; Huang, Z. Structural basis of assembly of the human T cell receptor–CD3 complex. Nature 2019, 573, 546–552. [Google Scholar] [CrossRef] [PubMed]
  133. Dushek, O.; Goyette, J.; Van Der Merwe, P.A. Non-catalytic tyrosine-phosphorylated receptors. Immunol. Rev. 2012, 250, 258–276. [Google Scholar] [CrossRef] [PubMed]
  134. Abram, C.L.; Lowell, C.A. The Expanding Role for ITAM-Based Signaling Pathways in Immune Cells. Sci. STKE 2007, 2007, re2. [Google Scholar] [CrossRef] [PubMed]
  135. Reddy, M.; Eirikis, E.; Davis, C.; Davis, H.M.; Prabhakar, U. Comparative analysis of lymphocyte activation marker expression and cytokine secretion profile in stimulated human peripheral blood mononuclear cell cultures: An in vitro model to monitor cellular immune function. J. Immunol. Methods 2004, 293, 127–142. [Google Scholar] [CrossRef] [PubMed]
  136. Bridgeman, J.S.; Hawkins, R.E.; Bagley, S.; Blaylock, M.; Holland, M.; Gilham, D.E. The Optimal Antigen Response of Chimeric Antigen Receptors Harboring the CD3ζ Transmembrane Domain Is Dependent upon Incorporation of the Receptor into the Endogenous TCR/CD3 Complex. J. Immunol. 2010, 184, 6938–6949. [Google Scholar] [CrossRef] [Green Version]
  137. Stoiber, S.; Cadilha, B.L.; Benmebarek, M.-R.; Lesch, S.; Endres, S.; Kobold, S. Limitations in the Design of Chimeric Antigen Receptors for Cancer Therapy. Cells 2019, 8, 472. [Google Scholar] [CrossRef] [Green Version]
  138. Woerly, G.; Lacy, P.; Ben Younes, A.; Roger, N.; Loiseau, S.; Moqbel, R.; Capron, M. Human eosinophils express and release IL-13 following CD28-dependent activation. J. Leukoc. Boil. 2002, 72, 769–779. [Google Scholar]
  139. Venuprasad, K.; Parab, P.; Prasad, D.V.R.; Sharma, S.; Banerjee, P.P.; Deshpande, M.; Mitra, D.K.; Pal, S.; Bhadra, R.; Mitra, D.; et al. Immunobiology of CD28 expression on human neutrophils. I. CD28 regulates neutrophil migration by modulating CXCR-1 expression. Eur. J. Immunol. 2001, 31, 1536–1543. [Google Scholar] [CrossRef]
  140. Williams, J.A.; Lumsden, J.M.; Yu, X.; Feigenbaum, L.; Zhang, J.; Steinberg, S.M.; Hodes, R.J. Regulation of thymic NKT cell development by the B7-CD28 costimulatory pathway. J. Immunol. 2008, 181, 907–917. [Google Scholar] [CrossRef] [Green Version]
  141. Michel, F.M.; Attal-Bonnefoy, G.; Mangino, G.; Mise-Omata, S.; Acuto, O. CD28 as a molecular amplifier extending TCR ligation and signaling capabilities. Immunity 2001, 15, 935–945. [Google Scholar] [CrossRef] [Green Version]
  142. Acuto, O.; Cantrell, D. T Cell Activation and the Cytoskeleton. Annu. Rev. Immunol. 2000, 18, 165–184. [Google Scholar] [CrossRef] [PubMed]
  143. Porciello, N.; Tuosto, L. CD28 costimulatory signals in T lymphocyte activation: Emerging functions beyond a qualitative and quantitative support to TCR signalling. Cytokine Growth Factor Rev. 2016, 28, 11–19. [Google Scholar] [CrossRef] [PubMed]
  144. Boomer, J.S.; Green, J.M. An Enigmatic Tail of CD28 Signaling. Cold Spring Harb. Perspect. Boil. 2010, 2, a002436. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  145. Milone, M.C.; Fish, J.D.; Carpenito, C.; Carroll, R.G.; Binder, G.K.; Teachey, D.; Samanta, M.; Lakhal, M.; Gloss, B.; Danet-Desnoyers, G.; et al. Chimeric Receptors Containing CD137 Signal Transduction Domains Mediate Enhanced Survival of T Cells and Increased Antileukemic Efficacy In Vivo. Mol. Ther. 2009, 17, 1453–1464. [Google Scholar] [CrossRef]
  146. Lawson, D.H.; Lee, S.; Zhao, F.; Tarhini, A.A.; Margolin, K.A.; Ernstoff, M.S.; Atkins, M.B.; Cohen, G.I.; Whiteside, T.L.; Butterfield, L.H.; et al. Randomized, Placebo-Controlled, Phase III Trial of Yeast-Derived Granulocyte-Macrophage Colony-Stimulating Factor (GM-CSF) Versus Peptide Vaccination Versus GM-CSF Plus Peptide Vaccination Versus Placebo in Patients With No Evidence of Disease After Complete Surgical Resection of Locally Advanced and/or Stage IV Melanoma: A Trial of the Eastern Cooperative Oncology Group–American College of Radiology Imaging Network Cancer Research Group (E4697). J. Clin. Oncol. 2015, 33, 4066–4076. [Google Scholar] [CrossRef]
  147. Hombach, A.; Holzinger, A.; Abken, H. The Weal and Woe of Costimulation in the Adoptive Therapy of Cancer with Chimeric Antigen Receptor (CAR)-Redirected T Cells. Curr. Mol. Med. 2013, 13, 1079–1088. [Google Scholar] [CrossRef]
  148. Sadelain, M.; Brentjens, R.; Rivière, I. The basic principles of chimeric antigen receptor design. Cancer Discov. 2013, 3, 388–398. [Google Scholar] [CrossRef] [Green Version]
  149. Redeker, A.; Arens, R. Improving Adoptive T Cell Therapy: The Particular Role of T Cell Costimulation, Cytokines, and Post-Transfer Vaccination. Front. Immunol. 2016, 7, 189. [Google Scholar] [CrossRef] [Green Version]
  150. Faitschuk, E.; Hombach, A.; Frenzel, L.P.; Wendtner, C.-M.; Abken, H. Chimeric antigen receptor T cells targeting Fc μ receptor selectively eliminate CLL cells while sparing healthy B cells. Blood 2016, 128, 1711–1722. [Google Scholar] [CrossRef] [Green Version]
  151. Weinkove, R.; George, P.; Dasyam, N.; McLellan, A.D. Selecting costimulatory domains for chimeric antigen receptors: Functional and clinical considerations. Clin. Transl. Immunol. 2019, 8, e1049. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  152. Kintz, H.; Nylen, E.; Barber, A. Inclusion of Dap10 or 4-1BB costimulation domains in the chPD1 receptor enhances anti-tumor efficacy of T cells in murine models of lymphoma and melanoma. Cell. Immunol. 2020, 351, 104069. [Google Scholar] [CrossRef] [PubMed]
  153. Zhang, T.; Wu, M.-R.; Sentman, C.L. An NKp30-based chimeric antigen receptor promotes T cell effector functions and antitumor efficacy in vivo. J. Immunol. 2012, 189, 2290–2299. [Google Scholar] [CrossRef] [PubMed]
  154. Krug, C.; Birkholz, K.; Paulus, A.; Schwenkert, M.; Schmidt, P.; Hoffmann, N.; Hombach, A.; Fey, G.; Abken, H.; Schuler, G.; et al. Stability and activity of MCSP-specific chimeric antigen receptors (CARs) depend on the scFv antigen-binding domain and the protein backbone. Cancer Immunol. Immunother. 2015, 64, 1623–1635. [Google Scholar] [CrossRef] [PubMed]
  155. Frigault, M.J.; Lee, J.; Basil, M.C.; Carpenito, C.; Motohashi, S.; Scholler, J.; Kawalekar, O.U.; Guedan, S.; Mcgettigan, S.E.; Posey, A.D.; et al. Identification of chimeric antigen receptors that mediate constitutive or inducible proliferation of T cells. Cancer Immunol. Res. 2015, 3, 356–367. [Google Scholar] [CrossRef] [Green Version]
  156. Lee, D.W.; Kochenderfer, J.N.; Stetler-Stevenson, M.; Cui, Y.K.; Delbrook, C.; A Feldman, S.; Fry, T.J.; Orentas, R.; Sabatino, M.; Shah, N.N.; et al. T cells expressing CD19 chimeric antigen receptors for acute lymphoblastic leukaemia in children and young adults: A phase 1 dose-escalation trial. Lancet 2015, 385, 517–528. [Google Scholar] [CrossRef]
  157. Park, J.H.; Riviere, I.; Gonen, M.; Wang, X.; Sénéchal, B.; Curran, K.J.; Sauter, C.; Wang, Y.; Santomasso, B.; Mead, E.; et al. Long-Term Follow-up of CD19 CAR Therapy in Acute Lymphoblastic Leukemia. N. Engl. J. Med. 2018, 378, 449–459. [Google Scholar] [CrossRef]
  158. Lang, S.; Vujanovic, N.L.; Wollenberg, B.; Whiteside, T.L. Absence of B7.1-CD28/CTLA-4-mediated co-stimulation in human NK cells. Eur. J. Immunol. 1998, 28, 780–786. [Google Scholar] [CrossRef]
  159. Goodier, M.R.; Londei, M. CD28 is not directly involved in the response of human CD3-CD56+ natural killer cells to lipopolysaccharide: A role for T cells. Immunology 2004, 111, 384–390. [Google Scholar] [CrossRef]
  160. Verma, I.M.; Van Antwerp, D.; Stevenson, J.K.; Schwarz, E.M.; Miyamoto, S. Rel/NF-kappa B/I kappa B family: Intimate tales of association and dissociation. Genes Dev. 1995, 9, 2723–2735. [Google Scholar] [CrossRef] [Green Version]
  161. Melero, I.; Johnston, J.V.; Shufford, W.W.; Mittler, R.S.; Chen, L. NK1.1 Cells Express 4-1BB (CDw137) Costimulatory Molecule and Are Required for Tumor Immunity Elicited by Anti-4-1BB Monoclonal Antibodies. Cell. Immunol. 1998, 190, 167–172. [Google Scholar] [CrossRef] [PubMed]
  162. Drenkard, D.; Becke, F.M.; Langstein, J.; Spruss, T.; Kunz-Schughart, L.A.; Tan, T.E.; Lim, Y.C.; Schwarz, H. CD137 is expressed on blood vessel walls at sites of inflammation and enhances monocyte migratory activity. FASEB J. 2006, 21, 456–463. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  163. Von Kempis, J.; Schwarz, H.; Lotz, M. Differentiation-dependent and stimulus-specific expression of ILA, the human 4-1BB-homologue, in cells of mesenchymal origin. Osteoarthr. Cartil. 1997, 5, 394–406. [Google Scholar] [CrossRef] [Green Version]
  164. Cannons, J.L.; Choi, Y.; Watts, T.H. Role of TNF receptor-associated factor 2 and p38 mitogen-activated protein kinase activation during 4-1BB-dependent immune response. J. Immunol. 2000, 165, 6193–6204. [Google Scholar] [CrossRef] [Green Version]
  165. Lee, H.-W.; Nam, K.-O.; Park, S.-J.; Kwon, B.S. 4-1BB enhances CD8+ T cell expansion by regulating cell cycle progression through changes in expression of cyclins D and E and cyclin-dependent kinase inhibitor p27kip1. Eur. J. Immunol. 2003, 33, 2133–2141. [Google Scholar] [CrossRef]
  166. Imai, C.; Iwamoto, S.; Campana, D. Genetic modification of primary natural killer cells overcomes inhibitory signals and induces specific killing of leukemic cells. Blood 2005, 106, 376–383. [Google Scholar] [CrossRef] [Green Version]
  167. Fraietta, J.A.; Lacey, S.F.; Orlando, E.J.; Pruteanu-Malinici, I.; Gohil, M.; Lundh, S.; Boesteanu, A.C.; Wang, Y.; O’Connor, R.S.; Hwang, W.-T.; et al. Determinants of response and resistance to CD19 chimeric antigen receptor (CAR) T cell therapy of chronic lymphocytic leukemia. Nat. Med. 2018, 24, 563–571. [Google Scholar] [CrossRef]
  168. Maude, S.L.; Laetsch, T.W.; Buechner, J.; Rives, S.; Boyer, M.; Bittencourt, H.; Bader, P.; Verneris, M.R.; Stefanski, H.E.; Myers, G.D.; et al. Tisagenlecleucel in Children and Young Adults with B-Cell Lymphoblastic Leukemia. N. Engl. J. Med. 2018, 378, 439–448. [Google Scholar] [CrossRef]
  169. Houchins, J.P.; Yabe, T.; McSherry, C.; Bach, F.H. DNA sequence analysis of NKG2, a family of related cDNA clones encoding type II integral membrane proteins on human natural killer cells. J. Exp. Med. 1991, 173, 1017–1020. [Google Scholar] [CrossRef] [Green Version]
  170. Bauer, S. Activation of NK Cells and T Cells by NKG2D, a Receptor for Stress-Inducible MICA. Science 1999, 285, 727–729. [Google Scholar] [CrossRef]
  171. Raulet, D.H. Roles of the NKG2D immunoreceptor and its ligands. Nat. Rev. Immunol. 2003, 3, 781–790. [Google Scholar] [CrossRef] [PubMed]
  172. González, S.; López-Soto, A.; Suárez-Álvarez, B.; López-Vázquez, A.; López-Soto, A.; Gonzalez, S. NKG2D ligands: Key targets of the immune response. Trends Immunol. 2008, 29, 397–403. [Google Scholar] [CrossRef] [PubMed]
  173. Zafirova, B.; Wensveen, F.M.; Gulin, M.; Polić, B. Regulation of immune cell function and differentiation by the NKG2D receptor. Cell. Mol. Life Sci. 2011, 68, 3519–3529. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  174. Serrano, A.E.; Menares-Castillo, E.; Garrido-Tapia, M.; Ribeiro, C.H.; Hernández, C.J.; Mendoza-Naranjo, A.; Gatica-Andrades, M.; Valenzuela-Diaz, R.; Zuñiga, R.; López, M.N.; et al. Interleukin 10 decreases MICA expression on melanoma cell surface. Immunol. Cell Boil. 2010, 89, 447–457. [Google Scholar] [CrossRef] [PubMed]
  175. Jamieson, A.M.; Diefenbach, A.; McMahon, C.W.; Xiong, N.; Carlyle, J.R.; Raulet, D.H. The Role of the NKG2D Immunoreceptor in Immune Cell Activation and Natural Killing. Immunity 2002, 17, 19–29. [Google Scholar] [CrossRef] [Green Version]
  176. Diefenbach, A.; Tomasello, E.; Lucas, M.; Jamieson, A.M.; Hsia, J.K.; Vivier, E.; Raulet, D.H. Selective associations with signaling proteins determine stimulatory versus costimulatory activity of NKG2D. Nat. Immunol. 2002, 3, 1142–1149. [Google Scholar] [CrossRef]
  177. Zhang, T.; Lemoi, B.A.; Sentman, C.L. Chimeric NK-receptor–bearing T cells mediate antitumor immunotherapy. Blood 2005, 106, 1544–1551. [Google Scholar] [CrossRef]
  178. Demoulin, B.; Cook, W.J.; Murad, J.; Graber, D.J.; Sentman, M.-L.; Lonez, C.; E Gilham, D.; Sentman, C.L.; Agaugue, S. Exploiting natural killer group 2D receptors for CAR T-cell therapy. Futur. Oncol. 2017, 13, 1593–1605. [Google Scholar] [CrossRef] [Green Version]
  179. Zhang, T.; Barber, A.; Sentman, C.L. Generation of Antitumor Responses by Genetic Modification of Primary Human T Cells with a Chimeric NKG2D Receptor. Cancer Res. 2006, 66, 5927–5933. [Google Scholar] [CrossRef] [Green Version]
  180. Chang, Y.-H.; Connolly, J.; Shimasaki, N.; Mimura, K.; Kono, K.; Campana, D. A Chimeric Receptor with NKG2D Specificity Enhances Natural Killer Cell Activation and Killing of Tumor Cells. Cancer Res. 2013, 73, 1777–1786. [Google Scholar] [CrossRef] [Green Version]
  181. Wood, L.D.; Parsons, D.W.; Jones, S.; Lin, J.; Sjöblom, T.; Leary, R.J.; Shen, N.; Boca, S.M.; Barber, T.; Ptak, J.; et al. The Genomic Landscapes of Human Breast and Colorectal Cancers. Science 2007, 318, 1108–1113. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  182. Barber, A.; Sentman, C.L. NKG2D receptor regulates human effector T-cell cytokine production. Blood 2011, 117, 6571–6581. [Google Scholar] [CrossRef] [Green Version]
  183. Zhang, T.; Sentman, C.L. Mouse tumor vasculature expresses NKG2D ligands and can be targeted by chimeric NKG2D-modified T cells. J. Immunol. 2013, 190, 2455–2463. [Google Scholar] [CrossRef] [Green Version]
  184. Lehner, M.; Gotz, G.; Proff, J.; Schaft, N.; Dorrie, J.; Full, F.; Ensser, A.; Muller, Y.A.; Cerwenka, A.; Abken, H.; et al. Redirecting T cells to Ewing’s sarcoma family of tumors by a chimeric NKG2D receptor expressed by lentiviral transduction or mRNA transfection. PLoS ONE 2012, 7, e31210. [Google Scholar] [CrossRef] [PubMed]
  185. Maasho, K.; Opoku-Anane, J.; Marusina, A.I.; Coligan, J.E.; Borrego, F. Cutting Edge: NKG2D Is a Costimulatory Receptor for Human Naive CD8+ T Cells. J. Immunol. 2005, 174, 4480–4484. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  186. Markiewicz, M.A.; Carayannopoulos, L.N.; Naidenko, O.V.; Matsui, K.; Burack, W.R.; Wise, E.L.; Fremont, D.H.; Allen, P.M.; Yokoyama, W.; Colonna, M.; et al. Costimulation through NKG2D enhances murine CD8+ CTL function: Similarities and differences between NKG2D and CD28 costimulation. J. Immunol. 2005, 175, 2825–2833. [Google Scholar] [CrossRef] [Green Version]
  187. Upshaw, J.L.; Leibson, P.J. NKG2D-mediated activation of cytotoxic lymphocytes: Unique signaling pathways and distinct functional outcomes. Semin. Immunol. 2006, 18, 167–175. [Google Scholar] [CrossRef]
  188. Lanier, L.L. DAP10- and DAP12-associated receptors in innate immunity. Immunol. Rev. 2009, 227, 150–160. [Google Scholar] [CrossRef] [Green Version]
  189. Rajasekaran, K.; Xiong, V.; Fong, L.; Gorski, J.; Malarkannan, S. Functional Dichotomy between NKG2D and CD28-Mediated Co-Stimulation in Human CD8+ T Cells. PLoS ONE 2010, 5, e12635. [Google Scholar] [CrossRef] [Green Version]
  190. Whitman, E.; Barber, A. NKG2D receptor activation of NF-κB enhances inflammatory cytokine production in murine effector CD8+ T cells. Mol. Immunol. 2015, 63, 268–278. [Google Scholar] [CrossRef]
  191. McQueen, B.; Trace, K.; Whitman, E.; Bedsworth, T.; Barber, A. Natural killer group 2D and CD28 receptors differentially activate mammalian/mechanistic target of rapamycin to alter murine effector CD8+T-cell differentiation. Immunology 2016, 147, 305–320. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  192. Lynch, A.; Hawk, W.; Nylen, E.; Ober, S.; Autin, P.; Barber, A. Adoptive transfer of murine T cells expressing a chimeric-PD1-Dap10 receptor as an immunotherapy for lymphoma. Immunology 2017, 152, 472–483. [Google Scholar] [CrossRef] [PubMed]
  193. Parriott, G.; Deal, K.; Crean, S.; Richardson, E.; Nylen, E.; Barber, A. T cells expressing a chimeric-PD1-Dap10-CD3zeta receptor reduce tumor burden in multiple murine syngeneic models of solid cancer. Immunology 2020. [Google Scholar] [CrossRef] [PubMed]
  194. Zhao, Y.; Qiao, G.; Wang, X.; Song, Y.; Zhou, X.; Jiang, N.; Zhou, L.; Huang, H.; Zhao, J.; Morse, M.A.; et al. Combination of DC/CIK adoptive T cell immunotherapy with chemotherapy in advanced non-small-cell lung cancer (NSCLC) patients: A prospective patients’ preference-based study (PPPS). Clin. Transl. Oncol. 2018, 21, 721–728. [Google Scholar] [CrossRef] [PubMed]
  195. Lv, J.; Zhao, R.; Wu, D.; Zheng, D.; Wu, Z.; Shi, J.; Wei, X.; Wu, Q.; Long, Y.; Lin, S.; et al. Mesothelin is a target of chimeric antigen receptor T cells for treating gastric cancer. J. Hematol. Oncol. 2019, 12, 18. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  196. Duong, C.P.; Westwood, J.A.; Yong, C.S.M.; Murphy, A.; Devaud, C.; John, L.B.; Darcy, P.K.; Kershaw, M. Engineering T Cell Function Using Chimeric Antigen Receptors Identified Using a DNA Library Approach. PLoS ONE 2013, 8, e63037. [Google Scholar] [CrossRef] [Green Version]
  197. Fisher, J.; Abramowski, P.; Don, N.D.W.; Flutter, B.; Capsomidis, A.; Cheung, G.W.-K.; Gustafsson, K.; Anderson, J. Avoidance of On-Target Off-Tumor Activation Using a Co-stimulation-Only Chimeric Antigen Receptor. Mol. Ther. 2017, 25, 1234–1247. [Google Scholar] [CrossRef] [Green Version]
  198. Ribas, A.; Lawrence, D.; Atkinson, V.; Agarwal, S.; Miller, W.H.; Carlino, M.S.; Fisher, R.; Long, G.V.; Hodi, F.S.; Tsoi, J.; et al. Combined BRAF and MEK inhibition with PD-1 blockade immunotherapy in BRAF-mutant melanoma. Nat. Med. 2019, 25, 936–940. [Google Scholar] [CrossRef]
  199. Marin, V.; Kakuda, H.; Dander, E.; Imai, C.; Campana, D.; Biondi, A.; D’Amico, G. Enhancement of the anti-leukemic activity of cytokine induced killer cells with an anti-CD19 chimeric receptor delivering a 4-1BB-ζ activating signal. Exp. Hematol. 2007, 35, 1388–1397. [Google Scholar] [CrossRef]
  200. Wu, J.; Song, Y.; Bakker, A.B.H.; Bauer, S.; Spies, T.; Lanier, L.L.; Phillips, J.H. An Activating Immunoreceptor Complex Formed by NKG2D and DAP10. Science 1999, 285, 730–732. [Google Scholar] [CrossRef]
  201. Billadeau, D.D.; Upshaw, J.L.; A Schoon, R.; Dick, C.J.; Leibson, P.J. NKG2D-DAP10 triggers human NK cell–mediated killing via a Syk-independent regulatory pathway. Nat. Immunol. 2003, 4, 557–564. [Google Scholar] [CrossRef]
  202. Biagi, E.; Marin, V.; Attianese, G.M.P.G.; Dander, E.; D’Amico, G.; Biondi, A. Chimeric T-cell receptors: New challenges for targeted immunotherapy in hematologic malignancies. Haematology 2007, 92, 381–388. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  203. Fridman, W.H. Fc receptors and immunoglobulin binding factors 1. FASEB J. 1991, 5, 2684–2690. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  204. Raghavan, M.; Bjorkman, P.J. Fc Receptors and Their Interactions with Immunoglobulins. Annu. Rev. Cell Dev. Boil. 1996, 12, 181–220. [Google Scholar] [CrossRef] [Green Version]
  205. Nimmerjahn, F.; Ravetch, J.V. Fcγ receptors as regulators of immune responses. Nat. Rev. Immunol. 2008, 8, 34–47. [Google Scholar] [CrossRef] [PubMed]
  206. Takai, T.; Ono, M.; Hikida, M.; Ohmori, H.; Ravetch, J.V. Augmented humoral and anaphylactic responses in FcγRII-deficient mice. Nature 1996, 379, 346–349. [Google Scholar] [CrossRef] [PubMed]
  207. Vivier, E.; Rochet, N.; Ackerly, M.; Petrini, J.; Levine, H.; Daley, J.; Anderson, P. Signaling function of reconstituted CD16: ζ: γ receptor complex isoforms. Int. Immunol. 1992, 4, 1313–1323. [Google Scholar] [CrossRef]
  208. Schumann, G.; Dasgupta, J.D. Specificity of signal transduction through CD16, TCR-CD3 and BCR receptor chains containing the tyrosine-associated activation motif. Int. Immunol. 1994, 6, 1383–1392. [Google Scholar] [CrossRef]
  209. Indik, Z.; Park, J.; Hunter, S.; Schreiber, A. The molecular dissection of Fc gamma receptor mediated phagocytosis. Blood 1995, 86, 4389–4399. [Google Scholar] [CrossRef] [Green Version]
  210. Zhang, Y.; Boesen, C.C.; Radaev, S.; Brooks, A.G.; Fridman, W.H.; Sautès-Fridman, C.; Sun, P.D. Crystal Structure of the Extracellular Domain of a Human FcγRIII. Immunity 2000, 13, 387–395. [Google Scholar] [CrossRef] [Green Version]
  211. Caratelli, S.; Sconocchia, G.; Arriga, R.; Coppola, A.; Lanzilli, G.; Lauro, D.; Venditti, A.; Del Principe, M.I.; Lococo, F.; Maurillo, L.; et al. FCγ Chimeric Receptor-Engineered T Cells: Methodology, Advantages, Limitations, and Clinical Relevance. Front. Immunol. 2017, 8, 457. [Google Scholar] [CrossRef] [PubMed]
  212. Sacchetti, B.; Botticelli, A.; Pierelli, L.; Nuti, M.; Alimandi, M. CAR-T with License to Kill Solid Tumors in Search of a Winning Strategy. Int. J. Mol. Sci. 2019, 20, 1903. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  213. Ochi, F.; Fujiwara, H.; Tanimoto, K.; Asai, H.; Miyazaki, Y.; Okamoto, S.; Mineno, J.; Kuzushima, K.; Shiku, H.; Barrett, J.; et al. Gene-Modified Human / -T Cells Expressing a Chimeric CD16-CD3 Receptor as Adoptively Transferable Effector Cells for Anticancer Monoclonal Antibody Therapy. Cancer Immunol. Res. 2014, 2, 249–262. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  214. Kudo, K.; Imai, C.; Lorenzini, P.; Kamiya, T.; Kono, K.; Davidoff, A.M.; Chng, W.J.; Campana, D. T Lymphocytes Expressing a CD16 Signaling Receptor Exert Antibody-Dependent Cancer Cell Killing. Cancer Res. 2013, 74, 93–103. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  215. Clémenceau, B.; Valsesia-Wittmann, S.; Jallas, A.-C.; Vivien, R.; Rousseau, R.; Marabelle, A.; Caux, C.; Vié, H. In Vitro and In Vivo Comparison of Lymphocytes Transduced with a Human CD16 or with a Chimeric Antigen Receptor Reveals Potential Off-Target Interactions due to the IgG2 CH2-CH3 CAR-Spacer. J. Immunol. Res. 2015, 2015, 1–13. [Google Scholar] [CrossRef] [Green Version]
  216. D’Aloia, M.M.; Caratelli, S.; Palumbo, C.; Battella, S.; Arriga, R.; Lauro, D.; Palmieri, G.; Sconocchia, G.; Alimandi, M. T lymphocytes engineered to express a CD16-chimeric antigen receptor redirect T-cell immune responses against immunoglobulin G–opsonized target cells. Cytotherapy 2016, 18, 278–290. [Google Scholar] [CrossRef]
  217. Tanaka, H.; Fujiwara, H.; Ochi, F.; Tanimoto, K.; Casey, N.; Okamoto, S.; Mineno, J.; Kuzushima, K.; Shiku, H.; Sugiyama, T.; et al. Development of Engineered T Cells Expressing a Chimeric CD16-CD3 Receptor to Improve the Clinical Efficacy of Mogamulizumab Therapy Against Adult T-Cell Leukemia. Clin. Cancer Res. 2016, 22, 4405–4416. [Google Scholar] [CrossRef] [Green Version]
  218. Rataj, F.; Jacobi, S.J.; Stoiber, S.; Asang, F.; Ogonek, J.; Tokarew, N.; Cadilha, B.L.; Van Puijenbroek, E.; Heise, C.; Duewell, P.; et al. High-affinity CD16-polymorphism and Fc-engineered antibodies enable activity of CD16-chimeric antigen receptor-modified T cells for cancer therapy. Br. J. Cancer 2018, 120, 79–87. [Google Scholar] [CrossRef] [Green Version]
  219. Arriga, R.; Caratelli, S.; Lanzilli, G.; Ottaviani, A.; Cenciarelli, C.; Sconocchia, T.; Spagnoli, G.C.; Iezzi, G.; Roselli, M.; Lauro, D.; et al. CD16-158-valine chimeric receptor T cells overcome the resistance of KRAS-mutated colorectal carcinoma cells to cetuximab. Int. J. Cancer 2019, 146, 2531–2538. [Google Scholar] [CrossRef]
  220. Caratelli, S.; Arriga, R.; Sconocchia, T.; Ottaviani, A.; Lanzilli, G.; Pastore, D.; Cenciarelli, C.; Venditti, A.; Del Principe, M.I.; Lauro, D.; et al. In vitro elimination of epidermal growth factor receptor-overexpressing cancer cells by CD32A-chimeric receptor T cells in combination with cetuximab or panitumumab. Int. J. Cancer 2019, 146, 236–247. [Google Scholar] [CrossRef]
  221. Lee, J.-Y.; Forman, J.P.; McNerney, M.E.; Stepp, S.; Kuppireddi, S.; Guzior, D.; Latchman, Y.E.; Sayegh, M.H.; Yagita, H.; Park, C.-K.; et al. Requirement of homotypic NK-cell interactions through 2B4(CD244)/CD48 in the generation of NK effector functions. Blood 2005, 107, 3181–3188. [Google Scholar] [CrossRef] [Green Version]
  222. Agresta, L.; Hoebe, K.H.N.; Janssen, E.M. The Emerging Role of CD244 Signaling in Immune Cells of the Tumor Microenvironment. Front. Immunol. 2018, 9, 2809. [Google Scholar] [CrossRef] [PubMed]
  223. Valiante, N.M.; Trinchieri, G. Identification of a novel signal transduction surface molecule on human cytotoxic lymphocytes. J. Exp. Med. 1993, 178, 1397–1406. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  224. A Garni-Wagner, B.; Purohit, A.; A Mathew, P.; Bennett, M.; Kumar, V. A novel function-associated molecule related to non-MHC-restricted cytotoxicity mediated by activated natural killer cells and T cells. J. Immunol. 1993, 151, 60–70. [Google Scholar] [PubMed]
  225. Altvater, B.; Landmeier, S.; Pscherer, S.; Temme, J.; Schweer, K.; Kailayangiri, S.; Campana, D.; Juergens, H.; Pule, M.; Rossig, C. 2B4 (CD244) signaling by recombinant antigen-specific chimeric receptors costimulates natural killer cell activation to leukemia and neuroblastoma cells. Clin. Cancer Res. 2009, 15, 4857–4866. [Google Scholar] [CrossRef] [Green Version]
  226. Altvater, B.; Landmeier, S.; Pscherer, S.; Temme, J.; Juergens, H.; Pule, M.; Rossig, C. 2B4 (CD244) signaling via chimeric receptors costimulates tumor-antigen specific proliferation and in vitro expansion of human T cells. Cancer Immunol. Immunother. 2009, 58, 1991–2001. [Google Scholar] [CrossRef]
  227. Chen, L.; Liu, Y.; Zheng, Y.; Xu, J.; Zhang, Y.; Liu, W.; Li, Z.; Huang, G.; Li, W. Furanodienone overcomes temozolomide resistance in glioblastoma through the downregulation of CSPG4-Akt-ERK signalling by inhibiting EGR1-dependent transcription. Phytotherapy Res. 2019, 33, 1736–1747. [Google Scholar] [CrossRef]
Figure 1. Schematic representation of all the CARs described in this review. Upper membrane: classical CAR models, lower two membranes: the more exotic CAR models.
Figure 1. Schematic representation of all the CARs described in this review. Upper membrane: classical CAR models, lower two membranes: the more exotic CAR models.
Ijms 21 03525 g001
Figure 2. Summarizing representation of the normal roles of potential CAR signaling domains. Arrows indicate how the signaling pathways of different surface receptors are interconnected respectively to find resemblance in other cell types. Golden bars represent SH3 binding motifs, light green bars ITSMs, red bars ITAMs. More details on the individual proteins are discussed in the text.
Figure 2. Summarizing representation of the normal roles of potential CAR signaling domains. Arrows indicate how the signaling pathways of different surface receptors are interconnected respectively to find resemblance in other cell types. Golden bars represent SH3 binding motifs, light green bars ITSMs, red bars ITAMs. More details on the individual proteins are discussed in the text.
Ijms 21 03525 g002
Table 1. List of clinical trials employing non-classical/alternative chimeric antigen receptor (CAR) vehicles or combinations listed on www.clinicaltrials.gov.
Table 1. List of clinical trials employing non-classical/alternative chimeric antigen receptor (CAR) vehicles or combinations listed on www.clinicaltrials.gov.
Cell TypeNotesTreatmentCancer TypesLocationsNCT Number 1
γ/δ T cellsNot yet recruitinganti-CD19-CAR γ/δ T cellsDifferent B-cell lymphomasBeijing, ChinaNCT02656147
Recruiting; observational onlydetermining number and viability of patients’ γ/δ T cells in preparation for a CAR trialAcute myeloid leukemia.Sutton, United KingdomNCT03885076
Not yet recruitingNKG2D-CAR γ/δ T cellsDifferent solid tumorsJohor Bahru, MalaysiaNCT04107142
NKT cellsWithdrawn, replaced by NCT03294954iC9-anti-GD2-CD28/OX40/CD3ζ CAR NKT (GINAKIT) cellsNeuroblastomaHouston, Texas, USANCT02439788
Recruiting; new CAR constructanti-GD2-CD28/CD3ζ-IL15 CAR NKT (GINAKIT2) cellsNeuroblastomaHouston, Texas, USANCT03294954
Not yet recruitinganti-CD19-CD28/CD3ζ-IL15 CAR NKT (ANCHOR) cellsDifferent B-cell lymphomasHouston, Texas, USANCT03774654
Combining CAR-T cells and DC vaccinationRecruitingCAR-T cells, engineered immune effector (EIE) CTLs, DCvac(no details available)NeurofibromatosisShenzhen, Guangdong, ChinaNCT04085159
RecruitingDifferent CAR-T cells, Eps8 or WT1 DCvacRelapsed/refractory Leukemia, myelodysplastic syndromeGuangzhou, Guangdong, ChinaNCT03291444
1 NCT number: CLINICALTRIALS.GOV IDENTIFIER.
Table 2. List of clinical trials employing CAR-NK cells listed on www.clinicaltrials.gov.
Table 2. List of clinical trials employing CAR-NK cells listed on www.clinicaltrials.gov.
NotesTreatmentCancer TypesLocationsNCT Number 1
Completedanti-CD19-4-1BB/CD3ζ
CAR-NK cells
Acute lymphoblastic leukemiaMemphis, Tennessee, USANCT00995137
Suspendedanti-CD19-CD3ζ/4-1BB
CAR-NK cells
B-cell acute lymphoblastic leukemiaSingaporeNCT01974479
Unknown statusanti-CD7-CD3ζ/CD28/4-1BB
CAR NK-92 cells
Acute myeloid leukemia and 8 more hematologic cancersSuzhou, Jiangsu, ChinaNCT02742727
Unknown statusanti-MUC1
CAR-NK cells
Hepatocellular carcinoma and 6 more solid cancersSuzhou, Jiangsu, ChinaNCT02839954
Unknown statusanti-CD19-TCRζ/CD28/4-1BB CAR-NK cellsAcute lymphocytic leukemia and 5 more hematologic cancersSuzhou, Jiangsu, ChinaNCT02892695
Unknown statusanti-CD33-CD3ζ/CD28/4-1BB CAR-NK cellsAcute myelogenous leukemia and 4 more hematologic cancersSuzhou, Jiangsu, ChinaNCT02944162
Recruiting; results publishedanti-CD19-CD28/CD3ζ/iCasp9/
IL-15 CAR-NK cells
Different B-cell lymphomasHouston, Texas, USANCT03056339
RecruitingNKG2D-CAR-NK cellsDifferent solid tumorsGuangzhou, Guangdong, ChinaNCT03415100
Not yet recruitinganti-PSMA
CAR-NK cells
Castration-resistant prostate cancerBeijing, ChinaNCT03692663
Not yet recruitinganti-CD19/CD22
CAR-NK cells
Refractory B-cell lymphomaBeijing, ChinaNCT03824964
Not yet recruitinganti-CD22
CAR-NK Cells
Refractory B-cell lymphomaBeijing, ChinaNCT03692767
Not yet recruitinganti-meso CAR-NK CellsEpithelial ovarian cancerBeijing, ChinaNCT03692637
Not yet recruitinganti-CD19
CAR-NK Cells
Refractory B-cell lymphomaBeijing, ChinaNCT03690310
Recruitinganti-BCMA CAR-NK-92 cellsMultiple myelomaWuxi, Jiangsu, ChinaNCT03940833
Recruitinganti-ROBO1 CAR-NK cellsPancreatic cancerShanghai, ChinaNCT03941457
Recruitinganti-ROBO1 CAR-NK cellsDifferent solid tumorsSuzhou, Jiangsu, ChinaNCT03940820
Withdrawn; lack of fundinganti-CD19-CD28/CD3ζ/iCasp9/IL-15 CAR-NK cellsDifferent B-cell lymphomasHouston, Texas, USANCT03579927
1 NCT number: CLINICALTRIALS.GOV IDENTIFIER.

Share and Cite

MDPI and ACS Style

Sievers, N.M.; Dörrie, J.; Schaft, N. CARs: Beyond T Cells and T Cell-Derived Signaling Domains. Int. J. Mol. Sci. 2020, 21, 3525. https://0-doi-org.brum.beds.ac.uk/10.3390/ijms21103525

AMA Style

Sievers NM, Dörrie J, Schaft N. CARs: Beyond T Cells and T Cell-Derived Signaling Domains. International Journal of Molecular Sciences. 2020; 21(10):3525. https://0-doi-org.brum.beds.ac.uk/10.3390/ijms21103525

Chicago/Turabian Style

Sievers, Nico M., Jan Dörrie, and Niels Schaft. 2020. "CARs: Beyond T Cells and T Cell-Derived Signaling Domains" International Journal of Molecular Sciences 21, no. 10: 3525. https://0-doi-org.brum.beds.ac.uk/10.3390/ijms21103525

Note that from the first issue of 2016, this journal uses article numbers instead of page numbers. See further details here.

Article Metrics

Back to TopTop