Abstract
Simple Summary
CAR T cell therapies have been successful at treating patients with blood cancers and are now FDA-approved therapies for various types of leukemias and lymphomas. However, there are many challenges to using CAR T cells to treat patients with solid tumors. In fact, no CAR T cell product is currently FDA-approved in this context. Optimism remains for this promising therapy to treat solid tumors in the future. Many different strategies are being employed to improve the efficacy of next-generation T cell products. This review focuses specifically on one key problem with current CAR T cell products for solid tumors: persistence. We describe the emerging strategies being used to improve this element of CAR T cell therapy.
Abstract
CAR T cells require optimization to be effective in patients with solid tumors. There are many barriers affecting their ability to succeed. One barrier is persistence, as to achieve an optimal antitumor response, infused CAR T cells must engraft and persist. This singular variable is impacted by a multitude of factors—the CAR T cell design, lymphodepletion regimen used, expansion method to generate the T cell product, and more. Additionally, external agents can be utilized to augment CAR T cells, such as the addition of novel cytokines, pharmaceutical drugs that bolster memory formation, or other agents during either the ex vivo expansion process or after CAR T cell infusion to support them in the oppressive tumor microenvironment. This review highlights many strategies being used to optimize T cell persistence as well as future directions for improving the persistence of infused cells.
Keywords: CAR T cells, adoptive cellular therapy, solid tumors, persistence
1. Introduction
Adoptive cellular transfer (ACT) therapy encompasses tumor-infiltrating lymphocyte (TIL) therapies as well as those that involve engineering T cells with either a T cell receptor (TCR) or chimeric antigen receptor (CAR). With TIL therapy, T cells from the tumor are expanded to large numbers before infusing them back into patients, whereas with both CAR and TCR-based therapies, the patient’s circulating T cells are obtained via leukapheresis and engineered with a receptor that can recognize a target expressed by the cancer [1]. CAR T cell therapy is particularly exciting and is decades in the making, with CAR T cells first described in the 1980s [2,3,4]. However, it was not until 2017 that the first CAR T cell therapy was FDA-approved—targeting CD19 on malignant B cells for use in treating patients with acute lymphoblastic leukemia (ALL) [5].
Since then, several additional CAR T cell therapies have been approved to treat patients with hematological malignancies, namely targeting CD19 and BCMA [6]. Current FDA-approved T cell therapies are listed in Table 1. This includes CAR T cell products for use in hematologic malignancies as well as the newly approved TIL product lifileucel and the TCR product afamitresgene autoleucel for use in melanoma and sarcoma, respectively. The TIL product lifileucel was approved in February 2024 for use in patients with melanoma unresponsive or refractory to immune checkpoint blockade therapy and was the first T cell therapy approved for use in solid tumors. Less than 6 months later, in August 2024, the second T cell therapy for solid tumors was approved for patients with unresectable or metastatic synovial sarcoma that have received chemotherapy previously, express the TCR-target antigen MAG121E-A4, and have one of the following HLA antigens: A*02:01P, -A*02:02P, -A*02:03P, or -A*02:06P [7]. While CAR T cell therapies are not yet approved for use in patients diagnosed with solid tumors, these rapid developments in this space highlight the potential future success of both CAR T and other T cell therapies in this context.
Table 1.
Company Name | Generic Name | Target |
---|---|---|
ABECMA | idecabtagene vicleucel | BCMA |
AMTAGVI | lifileucel | TIL for melanoma |
BREYANZI | lisocabtagene maraleucel | CD19 |
CARVYKTI | ciltacabtagene autoleucel | BCMA |
KYMRIAH | tisagenlecleucel | CD19 |
TECARTUS | brexucabtagene autoleucel | CD19 |
TECELRA | afamitresgene autoleucel | MAGE-A4 |
YESCARTA | axicabtagene ciloleucel | CD19 |
As demonstrated in Table 1, the approved CAR T cell products are for use in treating hematologic malignancies. Four products (Breyanzi, Kymriah, Tecartus, and Yescarta) target CD19, a cell surface marker expressed ubiquitously on B cells throughout development. Targeting CD19 via a CAR enables T cell products to more effectively treat B cell malignancies, directly recognizing a target expressed on malignant cells [8,9]. Abecma and Carvykti are additional CAR T cell products approved for the treatment of multiple myeloma, targeting the B cell maturation antigen (BCMA) expressed on malignant plasma cells in this cancer [10]. In contrast, many CAR T cell products designed for solid tumors target molecules that are expressed in both malignant and healthy tissues, increasing the risk of off-target effects, including organ damage. Additionally, a key difficulty in CAR T cell therapy for solid tumors is the necessity for CAR T cells to traffic into the site of the malignancies. This barrier is reduced when using CAR T cell therapies for hematologic malignancies, as they are able to effectively interact with their target cells while in circulation.
Both clinical and preclinical research suggests CAR T cells need to be further optimized to make an impact against solid tumors. Previous work has uncovered many reasons why CAR T cell therapy remains challenging for use in solid tumors, including (1) difficulty trafficking to tumors, (2) tumor heterogeneity, (3) T cell exhaustion, (4) decreased T cell persistence, (5) metabolic stress, (6) immunosuppressive tumor microenvironments, and (7) on-target off-tumor toxicities, among several other barriers reviewed elsewhere [11,12,13,14,15]. This new review focuses specifically on one of these barriers: persistence. Persistence describes the ability of T cells to survive long-term after infusion into patients and is associated with improved survival in hematological malignancies and importantly, in solid tumors [16,17]. Yet CAR T cell persistence in the context of solid tumors remains challenging, as they do not immediately encounter their CAR target and instead require time to traffic to the malignancy, during which they can encounter many stressors. Therefore, it is of utmost importance to improve CAR T cell persistence in patients with solid tumors.
This review seeks to define the plethora of ways in which translational scientists can enhance persistence as a means of improving CAR T cell efficacy in treating solid tumors. There are numerous points during the CAR T cell manufacturing process and after infusion into the patients, as displayed in Figure 1, that can be manipulated to augment persistence. This includes the initial design of the CAR T cell—such as what types of costimulatory domains are incorporated, the receptor affinity, and whether the CAR T cell has been engineered with additional attributes that will impact its ability to engraft and persist. Secondly, the conditions for expansion and level of stemness of the CAR T cell product are important contributors to persistence. Before patients receive these T cell products, they can undergo lymphodepleting regimens that greatly impact the ability of CAR T cells to engraft and persist. Lastly, after CAR T cell infusion into patients, additional agents or vaccination strategies can improve the persistence of adoptively transferred cells. These factors are discussed herein.
2. Discussion
2.1. CAR T Cell Design
The design of the CAR T cell construct is an important aspect to consider. CAR T cells are normally designed with an extracellular single-chain variable fragment (scFv), followed by the transmembrane domain, an intracellular region that contains the CD3ζ activation domain, and costimulatory domains [18]. These signaling elements differ depending on the generation of the CAR T cell, with the first generation only containing the CD3ζ domain, while the second and third generations have one or two costimulatory domains, respectively [18].
Depending on which costimulatory domain(s) are incorporated, CAR T cells can be endowed with different properties. Current FDA-approved CAR T cell therapies use either CD28 or 4-1BB (CD137) costimulatory domains. Canonically, CD28 is more associated with effector phenotypes, while 4-1BB is associated with enhanced persistence due to their different kinetics, although both have demonstrated efficacy in preclinical models and clinically in humans [19,20,21]. Additionally, when combined in a third-generation CAR, these two costimulatory domains led to more robust persistence in leukemia and solid tumor models [19,22]. Multiple third-generation CAR T cell products are in early-phase clinical trials, spanning the treatment of patients with thyroid cancer, neuroblastoma, and melanoma. Treatment with these products shows great promise, demonstrating CAR T persistence and disease response [23,24].
Several costimulatory domains can be incorporated into CAR constructs (Figure 2). These cues can be incorporated as the sole costimulatory domain (second-generation CAR) or in combination with additional costimulatory molecules (third-generation CAR). ICOS was reported to improve persistence and, when combined with 4-1BB in a third-generation CAR T construct, mediated superior antitumor effects against lung tumors [25]. CD2 has also been incorporated into MUC1-targeting CAR T constructs in a Phase I clinical trial for solid tumors [26]. Other novel CAR constructs have also been made, integrating MyD88 combined with CD40 to enhance CAR T persistence [27,28] or DNAX-activating proteins (DAP) 10 and 12 [29,30,31]. CD27 inclusion in CAR constructs also demonstrates potential benefit to CAR T survival [32].
There remains a question of which of these costimulatory domains and in what combination in the construct will lead to the best CAR T cell products for solid tumors. This uncertainty is in part due to making T cells with the “right” biology. In other words, while it is well appreciated that effector functions are important in CAR T success, the sustained signaling and activation that foster these properties can also lead to their dysfunction [33]. Sustained activation even in the absence of antigen stimulation is also defined as tonic signaling and can limit antitumor efficacy [33].
Strategies to ameliorate tonic signaling have been reviewed elsewhere [34] and are an important consideration when designing CAR constructs. Key factors influencing tonic signaling include the folding and stability of the single-chain variable fragment (scFv), spacer flexibility and length, target antigen affinity, and the choice of transmembrane and costimulatory domains [34]. DAP12 can be incorporated as a costimulatory domain to reduce tonic signaling due to its immunoreceptor tyrosine-based inhibitory motif (ITIM) [29]. The choice of scFv in CAR T cells is also crucial for success, as its stability can lead to self-aggregation and heightened tonic signaling [35]. Different “tonic signaling indices” for commonly used CARs were calculated using normalized CD69 levels [36]. This same group found variable levels of tonic signaling depending on the construct and that high levels of tonic signaling correlated with increased T cell exhaustion signatures [36]. As a result, several strategies are being employed to aid in tonic signaling effects, which are reviewed elsewhere [34].
Logic-gated strategies and dual-targeting CAR T cells offer exciting solutions for minimizing tonic signaling and off-target effects. Boolean logic gates—“AND,” “NOT,” and “OR”—enable CAR T cells to activate only in the presence of specific antigen combinations (AND), reduce activation in non-target cells (NOT), or respond to any of multiple molecules (OR) [37]. One unique logic-gated strategy involves using combinatorial antigen-sensing circuits, specifically SynNotch circuits. SynNotch receptors are designed so that an extracellular cue, such as initial binding to tumor antigen, can then trigger an engineered cellular response, such as the expression of an additional CAR [38]. In CAR T cells, this feature enables more precise CAR expression at tumor sites and reduces the tonic signaling typically observed with CAR products [38,39], allowing for localized CAR expression and reducing on-target, off-tumor effects [39]. SynNotch CAR T cells have displayed both increased persistence and antitumor activity in mice bearing solid tumors and represent a promising therapeutic strategy [40].
Herein, we review changes to CAR design that can augment their persistence in vivo; however, it is important to acknowledge that a plethora of other elements can be considered for novel CAR design to generally improve their efficacy in solid tumors. Some of these areas of ongoing research pertain to choosing the optimal target antigen, minimizing toxicities and off-target effects, and considering synapse formation and temporal dynamics of CAR signaling [18]. Furthermore, spacer length and receptor affinity are crucial elements known to impact CAR T cell efficacy [41]. Additionally, to combat novel barriers such as immunosuppressive microenvironments and trafficking through tissue, there may be other elements that need to be incorporated into CAR designs for therapy success in patients with solid tumors.
2.2. Genetic Alteration of CAR T Cells
The gene expression of the CAR-modified T cells can also be modulated to impact CAR therapy efficacy and persistence. This is primarily accomplished in two ways: overexpressing pathways to promote CAR T cell fitness or inactivating pathways that are deleterious for T cell therapy efficacy. Many genes have improved CAR T cell products when overexpressed. One recent strategy involved overexpressing the active form of the transcription factor FOXO1 (FOXO1-ADA) in CAR T cells, finding that when increased, CAR T cells had stemness and improved antitumor abilities in mice with solid tumors [42].
Similar strategies have been used to overexpress other important molecules in T cell fitness. Overexpression of Ado deaminase (ADA-OE), an enzyme important in metabolizing the immunosuppressive metabolite Ado, led to increased properties of stemness [43] and could be utilized in solid tumor models in the future. When overexpressed, c-Jun led to increased CAR T cell expansion as well as enhanced functionality in murine solid tumor models [44]. Metabolic mediators like GLUT1 and GSNOR have also been found to enhance CAR products when increased [45,46]. Persistence, specifically, was found to be improved by overexpression of the anti-apoptotic BCL-XL [47]. Chemokines can also be overexpressed to mediate the improved effects of CAR therapies [48]. There are a plethora of genes that could ultimately be upregulated in CAR T cells to mediate improved responses, and this strategy is yet another means to improve CAR therapies.
One strategy used to identify genes that may impact stemness was to screen for mutations found in T cell neoplasms associated with improved T cell fitness [49]. The effect of these mutations on CAR T cell phenotype and persistence was then assessed using pooled screening tools and a xenograft model. Specifically, CAR T cell persistence was increased when genes such as MYCN, CARD11/PIK3R3 fusion, CCND3, STAT3, and others were enriched [49]. CARD11–PIK3R3 was further investigated and found to improve CAR T cell efficacy in multiple solid tumor models as well as enhance their survival [49]. Tools such as this screening model can be used to help identify targets beneficial to T cell survival that can then be overexpressed to increase CAR T survival.
Conversely, inactivation of various pathways or enzymes can also enhance CAR T cell products. For example, groups have found that disruption of SUV39H1, a histone methyltransferase important in Th2 commitment [50], improves CAR T cell persistence and endows them with a more “stem-like phenotype” in both leukemia and solid tumor models [51,52]. PD-1, a known mediator of T cell inhibition via the PD1-PDL1 axis [53], has also been knocked out in CAR T cells both in preclinical models and clinical trials for solid tumors [54,55,56,57]. CRISPR/Cas9 has revolutionized our ability to modulate CAR therapies for patients [58], and its use to knockout other inhibitory molecules like CTLA-4 [59], Lag-3 [60], and other factors can improve current therapies. Ultimately, perturbing gene expression in CAR T cell products is a known strategy that can enhance the persistence and fitness of cells. This strategy is one to consider when optimizing their use for solid tumors.
2.3. Increasing T Cell Stemness
T cell stemness is an important aspect to consider for ACT therapy, as the infusion of more “memory-like” CD8 T cells has been correlated with improved persistence [61,62]. Decreased differentiation of T cells improves antitumor responses in solid tumors [63], and enhancing this attribute has been discussed as a major means to potentiate cellular therapy [64,65]. However, there does remain a balance between maintaining stemness and harnessing the cytolytic activity of T cells. There are many ways to increase stemness, including the initial selection of cells for CAR T cell transduction, the addition of cytokines, and drugs that can be added to culture during expansion.
Selecting T cells with naïve or stem-like properties can produce superior ACT products with distinct advantages in preclinical models [66,67]. Naïve CD8 T cells can be selected from bulk lymphocytes for transduction as one means to enhance CAR T therapies [68,69]. Likewise, a subset of CD4 cells, Th17 cells, have stem-like properties [70], and when redirected to express a CAR, they can lead to tumor regression in preclinical models [71,72]. Therefore, expanding cells with these polarizing cytokines or selecting them via extracellular markers is an additional way to improve CAR products. For instance, our team found increased expression of CD26 on CD4 T cells to mark a uniquely effective T cell population with enhanced migration, stemness, and persistence that may be of particular benefit for solid tumors [73,74]. These cells can be selected prior to transduction and expansion and have shown efficacy in CAR T models against hard-to-treat solid tumors [73,74].
Exogenous cytokines also impact T cell phenotypes. While the current most common approach is to expand cells in IL-2 (promoting T cell differentiation [75]), novel approaches using other cytokines have seen positive results [76]. IL-7 and IL-15 increase the survival and stemness of both CD4 and CD8 T cells [77,78,79]. When used during expansion, these cytokines can improve T cell stemness—increasing memory T cell populations that possess beneficial stem-like properties in addition to improved effector function in a model of B cell malignancy [61]. Similar strategies improve CAR T cell efficacy for solid tumors. One group found that by using a Naïve Pan T Cell collection kit (Miltenyi) to select for naïve T cells and subsequently activating them with CD3/28 and expanding with IL-7 and IL-15, there was a marked increase in stemness and improved antitumor activities [68]. Using these cytokines to augment CAR T cell products is a viable strategy to increase the stemness and persistence of ACT products. Additional cytokines can also enhance stemness features, including IL-24 [80] and IL-21 [81,82,83]. These may additionally add benefit to CAR T cells for solid tumors.
Other strategies to enhance stemness include using pharmacologic methods either during the expansion process or after infusion. The PI3-Kinase pathway in particular has been studied for its impact on T cell biology [84,85,86], and blockade of this pathway enhances ACT products [87,88,89]. This is at least in part mediated via increasing transcription factors in the Wnt/β-catenin pathway called Tcf1 and Lef1, associated with T cell stemness [88]. Various PI3K inhibitors have been reported to increase CAR T cell efficacy in leukemia models [90,91] and in TCR-based models for solid tumors [88,92,93,94]. Additional pharmacologic agents such as HDAC inhibitors [95], metformin [96], and mediators of Wnt signaling [97] enhance antitumor activity and may show promise for combining with CAR T cell therapy for solid tumors.
The costimulatory molecules used in T cell expansion and their potency are other variables often altered in the CAR T cell manufacturing process to improve phenotype. Beads or plate-bound antibodies are commonly used to activate T cells during the expansion process [76]. CD3/28 beads are most commonly used, but additional bead products such as CD3/ICOS beads can enhance certain CAR T cell products [71]. Moreover, the bead-to-T cell ratio using this CD3/ICOS strategy impacts CAR T responses as well and is yet another variable to consider [98]. Additional costimulatory molecules (CD137, CD2, CD40, etc.) have also been added to bead products to affect T cell phenotype and persistence, many of which are commercially available [99,100,101]. While the majority of current protocols utilize bead activation, others have used non-activated T cells as well [102].
Notably, other aspects need to be considered for generating the optimal CAR T cell for solid tumors. These factors include the expansion platform and level of oxygenation of the cells during manufacturing [103]. Transduction of the CAR construct is yet another factor that may impact their phenotype, function, and stemness. Modality, reagents used, and multiplicity of infection (MOI) are other factors to consider. The current FDA-approved products use lentiviral and retroviral methods [104]. However, non-viral methods also exist that can be used, such as the transposon systems [105,106,107]. CAR T cells engineered with the piggyBac transposon system have even been utilized in a clinical trial for non-small cell lung cancer patients [108], and this and other strategies may differentially impact T cell phenotype. Manufacturing strategies to improve CAR T success have been described elsewhere [109], encompassing how T cell activation, transduction, expansion, and other elements can impact the fate of CAR T cell products.
2.4. Cytokine Engineering
Many novel strategies to improve CAR T cell persistence involve cytokine engineering. The benefits of cytokines are described above in relation to their ability to enhance T cell stemness, yet there are additional cytokines helpful in augmenting CAR T cell persistence as well as unique strategies to increase the availability and potency of cytokines. One such agent includes rhIL-7-hyFc (efineptakin alfa, NT-I7), an interleukin-7 fusion protein that increases effector memory CAR T populations when injected into mice and improves overall CAR T persistence in mouse models [110].
As described previously, IL-15 can also increase the memory T cell phenotype, and there have been similar studies regarding its ability to benefit CAR T cell persistence. IL-15 can be administered systemically to enhance T cell persistence [111,112]; however, this can be associated with fever and rigors in part due to the marked expansion of natural killer (NK) cells [113]. Therefore, another strategy is to armor CAR T cells to produce cytokines, thereby decreasing systemic effects. CAR T cells engineered to produce membrane-tethered IL-15 alone or in combination with IL-21 led to enhanced persistence and antitumor responses [114,115]. Membrane-bound IL-15 (mbIL15) co-expressed with a second-generation CAR improved long-term engraftment even in the absence of tumor antigen [115]. IL-15 can also modulate the tumor microenvironment (TME) when secreted by CAR T cells to decrease myeloid-derived suppressor cells (MDSCs) in a glioblastoma (GBM) model [99]. These techniques aim to increase CAR T persistence and efficacy through cytokine modulation without the toxicities associated with systemic administration.
Unique strategies for delivering IL-2 via a CAR T cell product have also been investigated. This is particularly important, as systemic administration of IL-2 can lead to some toxicities [116,117,118]. To avoid these side effects, one group designed an orthogonal IL-2 and IL-2Rβ pair mutated in such a way as to prevent binding to wild-type human IL-2 [119]. This approach allowed for CAR T cells to be fitted with orthogonal IL-2Rβ, and upon administration of orthogonal IL-2 to CAR recipients, transferred CAR T cells were more specifically impacted while minimizing the systemic effects of IL-2 on host cells [119].
Beyond homeostatic cytokines, IL-12-secreting CAR T cells also show improvement in antitumor efficacy [120,121]. IL-12 has poor tolerability when administered systemically [122]. To abrogate this, CAR T cells can be “armored” to produce IL-12, allowing for more localized delivery. This strategy led to enhanced expansion and efficacy in mice with ovarian cancer as well as impacted tumor-associated macrophages [123], demonstrating how cytokines can impact the tumor microenvironment. It is likely that tumor microenvironments may respond differently to cytokines released by armored CAR T cells, depending on the malignancy. IL-12 can also be injected intratumorally [124] or as a plasmid/lipopolymer complex [125] for safer delivery. However, scientists have also begun armoring CAR T cells to produce other inflammatory cytokines in the IL-12 family, including IL-23.
IL-23-producing CAR T cells are particularly promising, able to regress neuroblastomas to a greater extent than IL-15- or IL-18-secreting CAR T cells [126]. Moreover, IL-23-producing CAR T cells had a better safety profile based on weight loss as a measurement [126]. Follow-up work, however, has shed a positive light on the impact of IL-18-producing CAR T cells to augment tumor immunity, which inspired a clinical trial testing the impact of IL-18 secretion by anti-CD19 CAR T cells in patients [127]. Along this line, CAR T cells armored to produce IL-18 also demonstrate preclinical benefits in solid tumors [128].
Several research groups have reported how a subset of cytokines, chemokines, can be utilized to enhance T cell homing and responses. Chemokines are directly relevant to CAR T cells, as their expression can mediate immune cell trafficking and their ability to migrate to and then infiltrate the tumor [129,130]. Particularly in the solid tumor context, CAR T cells can have difficulty penetrating the malignant tissue [131]. As different chemokines and chemokine receptors are expressed by tumors and have been correlated with infiltration and survival, this system can be hijacked to modulate CAR T cell homing towards the tumor [130,132,133]. For example, forced expression of CXCR6 improved the migration of CAR T cells into pancreatic tumors expressing CXCL16 in mice [134]. CXCR2-expressing cells also homed better to the tumors [135]. Clinical trials of CAR T cells combined with chemokine-based approaches are underway (NCT03602157, NCT05060796), and the role of these chemokines and chemokine ligands is an important aspect to consider in CAR T success.
Cytokines can shape both T cells and the surrounding tumor microenvironment. They can be administered systemically or can be produced by the CAR T cells themselves, as in the case of armored CARs. Especially in the context of solid tumors, where T cell survival and trafficking are of immense importance, cytokines play an important role. Cytokines can enhance CAR T cell persistence by promoting survival and growth, and they also play a crucial role in enabling effective migration to tumors. In closing, armoring CAR T cell therapies with distinct cytokines, chemokines, and other survival cues can ultimately improve their persistence, and all of the strategies discussed in this section have the potential to enhance CAR therapies for patients with solid tumors.
2.5. Decreased Expansion Time
Another very important aspect to consider is the expansion time of ACT products. CAR T cells are commonly expanded for around 9–14 days prior to infusion into patients, with some variability in this approach [136,137,138,139,140]. However, our lab has found that by decreasing this expansion time, we can deliver a superior ACT product [141]. Additional groups have similarly developed efficacious ACT products in a shorter time frame, finding that non-activated CAR T cells transduced and prepared within even 24 h have the potential to be efficacious for patients [102,142].
Importantly, by decreasing the time of ex vivo expansion, stemness properties can often be sustained or even enhanced, and clinical trials utilizing approaches to decreased expansion have shown promise [143]. CD8 T cell maturation was correlated with the time to expansion in one study [144], with decreased naïve T cells detected as expansion time was increased. However, it should be noted that the expansion platform used to generate ACT products can also affect these T cell phenotypes [103]. It should additionally be appreciated that by decreasing the expansion time, the final yield of CAR T cells may be compromised. Thus, it might be important to increase the number of T cells used during the manufacturing process. The enhanced T cell properties manifested by using a shorter expansion protocol must be balanced with this consideration. Considering these factors, we can optimize CAR T cell conditions for solid tumors and employ strategies to maintain stemness throughout development.
2.6. Lymphodepletion
One strategy to increase CAR T cell persistence is to precondition the patient with lymphodepletion prior to infusing the novel therapeutic product. Lymphodepletion directly augments the engraftment and persistence of transferred CAR T cells and TIL products, thereby improving long-term survival. Lymphodepletion can be mediated in several ways, ranging from treating the individual with radiation to chemotherapy-based regimens, both of which have been reported to improve ACT success in patients [145,146]. There are several mechanisms underlying the effectiveness of lymphodepletion: (1) increasing physiologic space for the transferred cells to thrive; (2) decreasing regulatory cell populations, including regulatory T cells (Tregs) and MDSCs, that blunt the expansion of infused T cells; (3) inducing immune activation of the innate immune system from microbial ligands; and (4) decreasing cytokine sinks, allowing for more available circulation of homeostatic cytokines (such as IL-7 and IL-15) that can directly promote the expansion and function of the transferred CAR T cells [146,147,148,149]. Moreover, lymphodepletion can increase the presentation of surface tumor antigens that can be presented to the infused T cells [150,151]. These benefits are visualized in Figure 3.
Specifically, CAR T cell therapy is augmented by lymphodepletion prior to CAR T cell transfer [152,153], with cyclophosphamide and fludarabine (alone or in combination) being the most common agents used to precondition the patient [154]. Additionally, in CD19-directed CARs, preconditioning patients with both cyclophosphamide and fludarabine improved T cell persistence [155]. We very recently reported that preconditioning with total body irradiation (TBI) or chemotherapy (cyclophosphamide/fludarabine alone or in combination) can differentially impact Th17 CD4 adoptive cellular therapy success, with some level of lymphodepletion needed for optimal efficacy [156]. Mechanistically, cytokines induced by lymphodepletion, such as MCP-1, are correlated with response; other cytokines, such as IL-7 and IL-15, are increased after lymphodepletion to aid T cell persistence and survival [153].
Importantly, though, patients with solid tumors may require differential methods of preconditioning compared to individuals diagnosed with hematologic malignancies prior to CAR T cell transfer. For example, in glioblastoma (GBM) patients, temozolomide increased CAR T cell persistence in the peripheral blood [157]. In another trial using an MPTK-CAR-T cell for mesothelin-positive tumors, cyclophosphamide and nab-paclitaxel with or without gemcitabine were used in the majority of patients [158]. Cyclophosphamide and fludarabine are still commonly used in the solid tumor context [159,160,161], yet they may not be the ideal candidates for each individual tumor type, and further investigation into these alternative regimens for preconditioning is still needed.
Lymphodepletion using either radiation or chemotherapy still has side effects for patients [154,162,163]—inspiring work by several groups to optimize CAR T cells to persist in the absence of lymphodepletion. This work includes strategies such as using CAR T cells that produce their own cytokines to augment persistence, such as with the engineered IL2 superkine (Super2) and IL33-producing CAR T cells [164] or IL-7 and CCL21-producing cells [165]. Another group identified STAT5b as a mediator of engraftment and used transient activation of this protein to improve transferred T cell persistence in lymphoreplete murine models [166]. These and other strategies may be able to decrease the need for lymphodepletion prior to cell transfer.
It is also possible that the type and level of lymphodepletion prior to CAR T cell therapy differ based on the patient’s diagnosed type of tumor. For example, CAR T cell therapy has shown some promise in treating intracranial brain tumors in the absence of lymphodepletion. In one case, multiple cycles of IL13Rα2-targeted CAR T cells were administered intracranially to a glioblastoma patient, leading to a complete response for 7.5 months before recurrence [167]. Another study involved intraventricular injections of CARv3-TEAM-E, targeting EGFRvIII and secreting T cell-engaging antibodies against wild-type EGFR, with mixed results: one patient had durable regression while the others had transient responses [168]. However, CAR T cell detection in cerebrospinal fluid (CSF) was limited between infusions.
In a different trial, glioblastoma patients received CART-EGFR-IL13Rα2 cells targeting both EGFR and IL13Rα2 intrathecally through an intraventricular reservoir, leading to a transient regression of the tumor in all patients and early persistence of CAR T cells in the CSF [169]. Locoregional IL-13Rα2-targeted CAR T cell therapy, including intratumoral and intraventricular injections, has shown promise for high-grade gliomas, suggesting that locoregional delivery may mitigate the need for lymphodepletion [170]. It is possible that this success is in part due to locoregional delivery of CAR T cells to the brain, as a study using intravenous administration of anti-EGFRvIII CAR T cells combined with pembrolizumab showed no clinical efficacy in lymphoreplete glioblastoma patients [171]. Moreover, little clinical impact was observed when patients were given cyclophosphamide and fludarabine before intravenous anti-EGFRvIII CAR T trials for glioblastoma [172]. Without a clinical trial that formally tests if lymphodepletion augments CAR T cells against brain cancer, it will be hard to untangle the impact of lymphodepletion on patient outcomes.
Given the distinct biology of intracranial tumors, such as the blood–brain barrier and immunosuppressive microenvironment, it is not surprising that CAR T cells have been successful in lymphoreplete patients in this context [173]. However, the optimal lymphodepletion regimen for solid tumors and its impact on CAR T cell therapies remain a critical question. Lymphodepletion may be necessary to achieve sustained, long-term results and could have beneficial effects on CAR T cell function and efficacy.
2.7. Immune Checkpoint Blockade and CAR T Cells in Combination
Immune checkpoint blockade (ICB) has undoubtedly changed the landscape of cancer therapies, with PD-1, CTLA-4, and PD-1/LAG-3 therapies becoming FDA-approved for use in multiple malignancies, leading to improved survival for many patients [174,175]. ICB works by blocking inhibitory T cell interactions [176] and therefore has implications for CAR T cell therapy. ICB can reinvigorate CAR T cells after infusion, as the expansion of CD19-targeting CAR T cells was seen in a patient given PD-1 blockade therapy [177]. Data regarding the combination of CAR T cells and ICB have been reviewed [178], and pembrolizumab combined with CAR T cells was found to be safe in Phase I clinical trials for several tumor types [171,179,180]. In the initial studies combining pembrolizumab and CAR T therapy for glioblastoma, efficacy was not demonstrated [171]. Yet ultimately, combinations of CAR T cells with PD-1 and other ICB agents for different solid tumor types remain to be further investigated.
CAR T cells express inhibitory markers after infusion into patients [181], and PD-1 blockade improves CAR T expansion in certain subsets of patients [182]. Treatment of malignant pleural disease with mesothelin-directed CAR T products followed by pembrolizumab bolstered their expansion and increased disease response, including prolonged stable disease [179]. However, both the timing of administration and how it is administered are likely to impact outcomes. Additionally, other ICBs may synergize with CAR T therapy, with current FDA-approved Lag-3 and CTLA-4 checkpoint blockade therapies available. Combination therapies with both ICBs and other agents are potential strategies to further improve current CAR T therapies and need further investigation regarding their efficacy.
2.8. Vaccination
Another way to enhance CAR T cell expansion and persistence involves vaccine strategies. Cancer vaccines use tumor antigens combined with adjuvants to help stimulate the immune system. They have shown some success in patients with many different types of solid tumors [183,184,185,186,187]. Importantly, however, in addition to their potential use as single agents to induce immune responses against tumors, vaccines hold great promise as an additive to other immunotherapies such as immune checkpoint blockade and adoptive cellular therapy. If combined with CAR T cells in particular, vaccines have the potential to improve the persistence and durability of these ACT products for solid tumors.
Such vaccination strategies have been used successfully in preclinical models as well as in a clinical trial (NCT04503278). One group used an RNA vaccine strategy combined with claudin 6-targeting CAR T cells [188]. They administered liposomal claudin 6-encoding RNA (CLDN6-LPX) after CAR T administration and detected enhanced CAR T persistence [188] and tolerability in patients administered these agents in a Phase I clinical trial [189]. Another novel strategy for vaccination involved administering a CAR T ligand with amphiphilic properties, termed amph-ligand, that can traffic to the lymph node to encounter and activate CAR T cells [190]. Others used dendritic cells loaded with the CAR T target antigen as a vaccination strategy to enhance therapy efficacy [191].
Vaccination is therefore another way to enhance CAR T cell persistence. Unlike hematologic malignancies, where CAR T cells more rapidly encounter their antigen after infusion, solid tumors may benefit from vaccination and CAR T combination strategies to enhance their function, in turn bolstering their expansion and persistence until they reach their target. There are many considerations for developing the optimal vaccine for this context, which have been reviewed by others [192,193], and this topic deserves additional consideration.
3. Conclusions
The persistence of CAR T cell products is an important mediator of therapy success and is a factor that deserves attention to improve these therapies for use in solid tumors. There are many unique challenges with solid tumors that can impact persistence. This includes immunosuppressive microenvironments, trafficking to the tumor site, tumor heterogeneity, and other barriers. However, despite these many barriers, we remain optimistic about the potential of CAR T cell therapies to benefit patients with a variety of solid tumor types. Herein, we reviewed a plethora of strategies that can modulate CAR T cell persistence. In summary, optimizing CAR T cell therapy involves several key factors. We comprehensively capture these factors visually in Figure 4, and these include: designing CAR T cells with costimulatory domains to enhance persistence or genetic modifications to boost survival; selecting or enhancing memory-like CAR T cells through cytokines or pharmacologic agents; ensuring effective expansion processes; considering lymphodepletion to improve CAR T cell engraftment and survival; and employing vaccination and combination with ICBs to promote CAR T cell persistence.
Many strategies can be taken to improve CAR T cell success. The question of which path will ultimately be the best to pursue remains. We ourselves have focused on both enhancing T cell stemness for ACT and optimizing lymphodepletion regimens prior to transfer [88,156]. Yet, undoubtedly, the CAR T cell design is another essential factor that will impact the field. Identifying factors that can enhance T cell trafficking and survival in the tumor is one way we can tailor their potency, and this can be improved in a multitude of ways.
In the grand scheme of therapies, CAR T cells are still a relatively new form of medicine. Their success in several hematologic malignancies is to be celebrated. Bringing these therapies to patients with solid tumors will require active effort, but we are fortunate as a field to have many minds working on this goal. Here, we describe a multitude of potential solutions to help improve these therapies. We are encouraged by the preclinical and clinical studies outlined herein that show promise for enhancing CAR T cell persistence.
As this review was limited to describing strategies that impact the specific topic of CAR T cell persistence, there were several topics not thoroughly discussed that are still important considerations. This includes (1) identifying the target antigen of interest [194], (2) modulating the tumor microenvironment [131], (3) aiding in T cell infiltration into solid tumors [195], and other challenges. Some solutions to improve T cell trafficking are described, such as modulating chemokine signaling, and there are reviews that delve into this topic [196,197,198]. There are also tumor-intrinsic factors to consider, such as (1) antigen expression, (2) location of the tumor, (3) cellular composition, and more. Brain tumors were specifically highlighted for several unique factors, but each solid tumor type has various aspects to consider when designing the optimal CAR T therapy. Yet, despite these challenges, T cell therapies are emerging as viable strategies for patients with solid tumors.
In this review, we summarized the strategies that can most directly improve CAR T cell persistence and, therefore, efficacy in solid tumors. There are many strategies considered, and a focus on several of these will likely yield the most optimal results. While there is a need for continued improvement of CAR T cell therapies, the ongoing research and novel strategies described herein demonstrate the progress being made towards CAR T cell success in solid tumors.
Author Contributions
Conceptualization, M.C.W. and C.M.P.; literature review, M.C.W. and K.G.D.; writing—original draft preparation, M.C.W.; writing—review and editing, C.M.P., A.C.C., B.B., M.C.W., and N.C.S.; supervision, C.M.P. All authors have read and agreed to the published version of the manuscript.
Conflicts of Interest
The authors disclose no conflicts of interest in relation to the published work. C.M.P. has previously received funds for consultancies/advisory boards/research contracts from Ares Immunotherapy, Lycera, Obsidian, and Thermo Fisher.
Funding Statement
This work was supported by the Melanoma Research Foundation (to MCW), NCI T32 CA272392-01A1 (to MCW), NCI F31CA281258-01A1 (to ACC), ARCs Foundation (to ACC), NCI R01CA287866-01, R01CA275199-02, V Foundation, plus MUSC and Emory University Start Up Funds (to CP).
Footnotes
Disclaimer/Publisher’s Note: The statements, opinions and data contained in all publications are solely those of the individual author(s) and contributor(s) and not of MDPI and/or the editor(s). MDPI and/or the editor(s) disclaim responsibility for any injury to people or property resulting from any ideas, methods, instructions or products referred to in the content.
References
- 1.Albarrán V., Román M.S., Pozas J., Chamorro J., Rosero D.I., Guerrero P., Calvo J.C., González C., de Quevedo C.G., de Aguado P.P., et al. Adoptive T cell therapy for solid tumors: Current landscape and future challenges. Front. Immunol. 2024;15:1352805. doi: 10.3389/fimmu.2024.1352805. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 2.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. doi: 10.1073/pnas.90.2.720. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 3.Gross G., Waks T., Eshhar Z. Expression of immunoglobulin-T-cell receptor chimeric molecules as functional receptors with antibody-type specificity. Proc. Natl. Acad. Sci. USA. 1989;86:10024–10028. doi: 10.1073/pnas.86.24.10024. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 4.Kuwana Y., Asakura Y., Utsunomiya N., Nakanishi M., Arata Y., Itoh S., Nagase F., Kurosawa Y. Expression of chimeric receptor composed of immunoglobulin-derived V regions and T-cell receptor-derived C regions. Biochem. Biophys. Res. Commun. 1987;149:960–968. doi: 10.1016/0006-291X(87)90502-X. [DOI] [PubMed] [Google Scholar]
- 5.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. doi: 10.1056/NEJMoa1709866. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 6.Mitra A., Barua A., Huang L., Ganguly S., Feng Q., He B. From bench to bedside: The history and progress of CAR T cell therapy. Front. Immunol. 2023;14:1188049. doi: 10.3389/fimmu.2023.1188049. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 7.FDA Approves First Gene Therapy to Treat Adults with Metastatic Synovial Sarcoma. [(accessed on 5 August 2024)];2024 Available online: https://www.fda.gov/news-events/press-announcements/fda-approves-first-gene-therapy-treat-adults-metastatic-synovial-sarcoma#:~:text=The%20product%20is%20an%20autologous,cancer%20cells%20in%20synovial%20sarcoma.
- 8.Sadelain M. CAR therapy: The CD19 paradigm. J. Clin. Investig. 2015;125:3392–3400. doi: 10.1172/JCI80010. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 9.Savoldo B., Grover N., Dotti G. CAR T cells for hematological malignancies. J. Clin. Investig. 2024;134:e177160. doi: 10.1172/JCI177160. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 10.Munshi N.C., Anderson L.D., Jr., Shah N., Madduri D., Berdeja J., Lonial S., Raje N., Lin Y., Siegel D., Oriol A., et al. Idecabtagene Vicleucel in Relapsed and Refractory Multiple Myeloma. N. Engl. J. Med. 2021;384:705–716. doi: 10.1056/NEJMoa2024850. [DOI] [PubMed] [Google Scholar]
- 11.Sorkhabi A.D., Khosroshahi L.M., Sarkesh A., Mardi A., Aghebati-Maleki A., Aghebati-Maleki L., Baradaran B. The current landscape of CAR T-cell therapy for solid tumors: Mechanisms, research progress, challenges, and counterstrategies. Front. Immunol. 2023;14:1113882. doi: 10.3389/fimmu.2023.1113882. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 12.Flugel C.L., Majzner R.G., Krenciute G., Dotti G., Riddell S.R., Wagner D.L., Abou-El-Enein M. Overcoming on-target, off-tumour toxicity of CAR T cell therapy for solid tumours. Nat. Rev. Clin. Oncol. 2023;20:49–62. doi: 10.1038/s41571-022-00704-3. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 13.Albelda S.M. CAR T cell therapy for patients with solid tumours: Key lessons to learn and unlearn. Nat. Rev. Clin. Oncol. 2023;21:47–66. doi: 10.1038/s41571-023-00832-4. [DOI] [PubMed] [Google Scholar]
- 14.Peng J.J., Wang L., Li Z., Ku C.L., Ho P.C. Metabolic challenges and interventions in CAR T cell therapy. Sci. Immunol. 2023;8:eabq3016. doi: 10.1126/sciimmunol.abq3016. [DOI] [PubMed] [Google Scholar]
- 15.Knochelmann H.M., Smith A.S., Dwyer C.J., Wyatt M.M., Mehrotra S., Paulos C.M. CAR T Cells in Solid Tumors: Blueprints for Building Effective Therapies. Front. Immunol. 2018;9:1740. doi: 10.3389/fimmu.2018.01740. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 16.Louis C.U., Savoldo B., Dotti G., Pule M., Yvon E., Myers G.D., Rossig C., Russell H.V., Diouf O., Liu E., et al. Antitumor activity and long-term fate of chimeric antigen receptor-positive T cells in patients with neuroblastoma. Blood. 2011;118:6050–6056. doi: 10.1182/blood-2011-05-354449. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 17.Porter D.L., Hwang W.-T., Frey N.V., Lacey S.F., Shaw P.A., Loren A.W., Bagg A., Marcucci K.T., Shen A., Gonzalez V., et al. Chimeric antigen receptor T cells persist and induce sustained remissions in relapsed refractory chronic lymphocytic leukemia. Sci. Transl. Med. 2015;7:303ra139. doi: 10.1126/scitranslmed.aac5415. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 18.Srivastava S., Riddell S.R. Engineering CAR-T cells: Design concepts. Trends Immunol. 2015;36:494–502. doi: 10.1016/j.it.2015.06.004. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 19.Zhao Z., Condomines M., Van Der Stegen S.J.C., 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. doi: 10.1016/j.ccell.2015.09.004. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 20.Sun C., Shou P., Du H., Hirabayashi K., Chen Y., Herring L.E., Ahn S., Xu Y., Suzuki K., Li G., et al. THEMIS-SHP1 Recruitment by 4-1BB Tunes LCK-Mediated Priming of Chimeric Antigen Receptor-Redirected T Cells. Cancer Cell. 2020;37:216–225 e6. doi: 10.1016/j.ccell.2019.12.014. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 21.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. doi: 10.1111/imr.12131. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 22.Hirabayashi K., Du H., Xu Y., Shou P., Zhou X., Fucá G., Landoni E., Sun C., Chen Y., Savoldo B., et al. Dual Targeting CAR-T Cells with Optimal Costimulation and Metabolic Fitness enhance Antitumor Activity and Prevent Escape in Solid Tumors. Nat. Cancer. 2021;2:904–918. doi: 10.1038/s43018-021-00244-2. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 23.Srour S.A., Villaflor V.M., Lorch J.H., Zafereo M.E., Gupta S., Hu M.I.-N., Dadu R., Lin A.Y., Lu Y., Ackatz L., et al. Safety and efficacy of AIC100 chimeric antigen receptor (CAR) T-cell therapy in patients with advanced thyroid cancers: Results from the phase 1 study. J. Clin. Oncol. 2024;42((Suppl. S16)):6112. [Google Scholar]
- 24.Del Bufalo F., De Angelis B., Caruana I., Del Baldo G., De Ioris M.A., Serra A., Mastronuzzi A., Cefalo M.G., Pagliara D., Amicucci M., et al. GD2-CART01 for Relapsed or Refractory High-Risk Neuroblastoma. N. Engl. J. Med. 2023;388:1284–1295. doi: 10.1056/NEJMoa2210859. [DOI] [PubMed] [Google Scholar]
- 25.Guedan S., Posey A.D., Jr., 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:e96976. doi: 10.1172/jci.insight.96976. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 26.Gutierrez R., Shah P.D., Hamid O., Garfall A.L., Posey A., Russell Bishop M., Blumenschein G.R., Lynne Johnson M., Lee S., Luke J.J., et al. Phase I experience with first in class TnMUC1 targeted chimeric antigen receptor T-cells in patients with advanced TnMUC1 positive solid tumors. J. Clin. Oncol. 2021;39((Suppl. S15)):e14513. doi: 10.1200/JCO.2021.39.15_suppl.e14513. [DOI] [Google Scholar]
- 27.Collinson-Pautz M.R., Chang W.-C., Lu A., Khalil M., Crisostomo J.W., Lin P.-Y., Mahendravada A., Shinners N.P., Brandt M.E., Zhang M., et al. Constitutively active MyD88/CD40 costimulation enhances expansion and efficacy of chimeric antigen receptor T cells targeting hematological malignancies. Leukemia. 2019;33:2195–2207. doi: 10.1038/s41375-019-0417-9. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 28.Prinzing B., Schreiner P., Bell M., Fan Y., Krenciute G., Gottschalk S. MyD88/CD40 signaling retains CAR T cells in a less differentiated state. JCI Insight. 2020;5:e136093. doi: 10.1172/jci.insight.136093. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 29.Zheng L., Ren L., Kouhi A., Khawli L.A., Hu P., Kaslow H.R., Epstein A.L. A Humanized Lym-1 CAR with Novel DAP10/DAP12 Signaling Domains Demonstrates Reduced Tonic Signaling and Increased Antitumor Activity in B-Cell Lymphoma Models. Clin. Cancer Res. 2020;26:3694–3706. doi: 10.1158/1078-0432.CCR-19-3417. [DOI] [PubMed] [Google Scholar]
- 30.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. doi: 10.1158/0008-5472.CAN-12-3558. [DOI] [PubMed] [Google Scholar]
- 31.Sun M., Xu P., Wang E., Zhou M., Xu T., Wang J., Wang Q., Wang B., Lu K., Wang C., et al. Novel two-chain structure utilizing KIRS2/DAP12 domain improves the safety and efficacy of CAR-T cells in adults with r/r B-ALL. Mol. Ther. Oncolytics. 2021;23:96–106. doi: 10.1016/j.omto.2021.08.014. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 32.Song D.G., Ye Q., Poussin M., Harms G.M., Figini M., Powell D.J., Jr. CD27 costimulation augments the survival and antitumor activity of redirected human T cells in vivo. Blood. 2012;119:696–706. doi: 10.1182/blood-2011-03-344275. [DOI] [PubMed] [Google Scholar]
- 33.Long A.H., Haso W.M., Shern J.F., Wanhainen K.M., 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. doi: 10.1038/nm.3838. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 34.Ajina A., Maher J. Strategies to Address Chimeric Antigen Receptor Tonic Signaling. Mol. Cancer Ther. 2018;17:1795–1815. doi: 10.1158/1535-7163.MCT-17-1097. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 35.Landoni E., Fucá G., Wang J., Chirasani V.R., Yao Z., Dukhovlinova E., Ferrone S., Savoldo B., Hong L.K., Shou P., et al. Modifications to the Framework Regions Eliminate Chimeric Antigen Receptor Tonic Signaling. Cancer Immunol. Res. 2021;9:441–453. doi: 10.1158/2326-6066.CIR-20-0451. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 36.Chen J., Qiu S., Li W., Wang K., Zhang Y., Yang H., Liu B., Li G., Li L., Chen M., et al. Tuning charge density of chimeric antigen receptor optimizes tonic signaling and CAR-T cell fitness. Cell Res. 2023;33:341–354. doi: 10.1038/s41422-023-00789-0. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 37.Han X., Wang Y., Wei J., Han W. Multi-antigen-targeted chimeric antigen receptor T cells for cancer therapy. J. Hematol. Oncol. 2019;12:128. doi: 10.1186/s13045-019-0813-7. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 38.Morsut L., Roybal K.T., Xiong X., Gordley R.M., Coyle S.M., Thomson M., Lim W.A. Engineering Customized Cell Sensing and Response Behaviors Using Synthetic Notch Receptors. Cell. 2016;164:780–791. doi: 10.1016/j.cell.2016.01.012. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 39.Roybal K.T., Rupp L.J., Morsut L., Walker W.J., McNally K.A., Park J.S., Lim W.A. Precision Tumor Recognition by T Cells with Combinatorial Antigen-Sensing Circuits. Cell. 2016;164:770–779. doi: 10.1016/j.cell.2016.01.011. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 40.Hyrenius-Wittsten A., Su Y., Park M., Garcia J.M., Alavi J., Perry N., Montgomery G., Liu B., Roybal K.T. SynNotch CAR circuits enhance solid tumor recognition and promote persistent antitumor activity in mouse models. Sci. Transl. Med. 2021;13:eabd8836. doi: 10.1126/scitranslmed.abd8836. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 41.Hudecek M., Lupo-Stanghellini M.-T., Kosasih P.L., Sommermeyer D., Jensen M.C., Rader C., Riddell S.R. Receptor affinity and extracellular domain modifications affect tumor recognition by ROR1-specific chimeric antigen receptor T cells. Clin. Cancer Res. 2013;19:3153–3164. doi: 10.1158/1078-0432.CCR-13-0330. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 42.Chan J.D., Scheffler C.M., Munoz I., Sek K., Lee J.N., Huang Y.-K., Yap K.M., Saw N.Y.L., Li J., Chen A.X.Y., et al. FOXO1 enhances CAR T cell stemness, metabolic fitness and efficacy. Nature. 2024;629:201–210. doi: 10.1038/s41586-024-07242-1. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 43.Klysz D.D., Fowler C., Malipatlolla M., Stuani L., Freitas K.A., Chen Y., Meier S., Daniel B., Sandor K., Xu P., et al. Inosine Induces Stemness Features in CAR T cells and Enhances Potency. Cancer Cell. 2024;42:266–282.e8. doi: 10.1016/j.ccell.2024.01.002. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 44.Lynn R.C., Weber E.W., Sotillo E., Gennert D., Xu P., Good Z., Anbunathan H., Lattin J., Jones R., Tieu V., et al. c-Jun overexpression in CAR T cells induces exhaustion resistance. Nature. 2019;576:293–300. doi: 10.1038/s41586-019-1805-z. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 45.Niu Q., Zhang H., Wang F., Xu X., Luo Y., He B., Shi M., Jiang E., Feng X. GSNOR overexpression enhances CAR-T cell stemness and anti-tumor function by enforcing mitochondrial fitness. Mol. Ther. 2024;32:1875–1894. doi: 10.1016/j.ymthe.2024.03.033. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 46.Shi Y., Kotchetkov I.S., Dobrin A., Hanina S.A., Rajasekhar V.K., Healey J.H., Sadelain M. GLUT1 overexpression enhances CAR T cell metabolic fitness and anti-tumor efficacy. Mol. Ther. 2024;32:2393–2405. doi: 10.1016/j.ymthe.2024.05.006. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 47.Korell F., Olson M.L., Salas-Benito D., Leick M.B., Larson R.C., Bouffard A., Silva H., Gasparetto A., Berger T.R., Kann M.C., et al. Comparative analysis of Bcl-2 family protein overexpression in CAR T cells alone and in combination with BH3 mimetics. Sci. Transl. Med. 2024;16:eadk7640. doi: 10.1126/scitranslmed.adk7640. [DOI] [PubMed] [Google Scholar]
- 48.Hu J.F., Wang Z.-W., Liao C.-Y., Chen Z.-W., Kang F.-P., Lin C.-F., Lin T.-S., Huang L., Tian Y.-F., Chen S. Induced expression of CCL19 promotes the anti-tumor ability of CAR-T cells by increasing their infiltration ability. Front. Immunol. 2022;13:958960. doi: 10.3389/fimmu.2022.958960. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 49.Hu J.-F., Wang Z.-W., Liao C.-Y., Chen Z.-W., Kang F.-P., Lin C.-F., Lin T.-S., Huang L., Tian Y.-F., Chen S. Naturally occurring T cell mutations enhance engineered T cell therapies. Nature. 2024;626:626–634. doi: 10.1038/s41586-024-07018-7. [DOI] [PubMed] [Google Scholar]
- 50.Allan R.S., Zueva E., Cammas F., Schreiber H.A., Masson V., Belz G.T., Roche D., Maison C., Quivy J.-P., Almouzni G., et al. An epigenetic silencing pathway controlling T helper 2 cell lineage commitment. Nature. 2012;487:249–253. doi: 10.1038/nature11173. [DOI] [PubMed] [Google Scholar]
- 51.Jain N., Zhao Z., Koche R.P., Antelope C., Gozlan Y., Montalbano A., Brocks D., Lopez M., Dobrin A., Shi Y., et al. Disruption of SUV39H1-Mediated H3K9 Methylation Sustains CAR T-cell Function. Cancer Discov. 2024;14:142–157. doi: 10.1158/2159-8290.CD-22-1319. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 52.López-Cobo S., Fuentealba J.R., Gueguen P., Bonté P.-E., Tsalkitzi K., Chacón I., Glauzy S., Bohineust A., Biquand A., Silva L., et al. SUV39H1 Ablation Enhances Long-term CAR T Function in Solid Tumors. Cancer Discov. 2024;14:120–141. doi: 10.1158/2159-8290.CD-22-1350. [DOI] [PubMed] [Google Scholar]
- 53.Parry R.V., Chemnitz J.M., Frauwirth K.A., Lanfranco A.R., Braunstein I., Kobayashi S.V., Linsley P.S., Thompson C.B., Riley J.L. CTLA-4 and PD-1 receptors inhibit T-cell activation by distinct mechanisms. Mol. Cell Biol. 2005;25:9543–9553. doi: 10.1128/MCB.25.21.9543-9553.2005. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 54.Lin Y., Chen S., Zhong S., An H., Yin H., Mcgowan E. 35O—Phase I clinical trial of PD-1 knockout anti-MUC1 CAR-T cells in the treatment of patients with non-small cell lung cancer. Ann. Oncol. 2019;30:xi12. [Google Scholar]
- 55.McGowan E., Lin Q., Ma G., Yin H., Chen S., Lin Y. PD-1 disrupted CAR-T cells in the treatment of solid tumors: Promises and challenges. Biomed. Pharmacother. 2020;121:109625. doi: 10.1016/j.biopha.2019.109625. [DOI] [PubMed] [Google Scholar]
- 56.Guo X., Jiang H., Shi B., Zhou M., Zhang H., Shi Z., Du G., Luo H., Wu X., Wang Y., et al. Disruption of PD-1 Enhanced the Anti-tumor Activity of Chimeric Antigen Receptor T Cells Against Hepatocellular Carcinoma. Front. Pharmacol. 2018;9:1118. doi: 10.3389/fphar.2018.01118. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 57.Chen S., Lin Y. Phase I clinical trial using a unique immunotherapeutic combination of MUC1-targeted CAR-T cells with PD-1-knockout in the treatment of patients with advanced esophageal cancer. J. Clin. Oncol. 2023;41((Suppl. S16)):e16061. doi: 10.1200/JCO.2023.41.16_suppl.e16061. [DOI] [Google Scholar]
- 58.Mirzaei H.R., Pourghadamyari H., Rahmati M., Mohammadi A., Nahand J.S., Rezaei A., Mirzaei H., Hadjati J. Gene-knocked out chimeric antigen receptor (CAR) T cells: Tuning up for the next generation cancer immunotherapy. Cancer Lett. 2018;423:95–104. doi: 10.1016/j.canlet.2018.03.010. [DOI] [PubMed] [Google Scholar]
- 59.Agarwal S., Aznar M.A., Rech A.J., Good C.R., Kuramitsu S., Da T., Gohil M., Chen L., Hong S.-J.A., Ravikumar P., et al. Deletion of the inhibitory co-receptor CTLA-4 enhances and invigorates chimeric antigen receptor T cells. Immunity. 2023;56:2388–2407 e9. doi: 10.1016/j.immuni.2023.09.001. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 60.Zhang Y., Zhang X., Cheng C., Mu W., Liu X., Li N., Wei X., Liu X., Xia C., Wang H. CRISPR-Cas9 mediated LAG-3 disruption in CAR-T cells. Front. Med. 2017;11:554–562. doi: 10.1007/s11684-017-0543-6. [DOI] [PubMed] [Google Scholar]
- 61.Xu Y., Zhang M., Ramos C.A., Durett A., Liu E., Dakhova O., Liu H., Creighton C.J., Gee A.P., Heslop H.E., et al. Closely related T-memory stem cells correlate with in vivo expansion of CAR.CD19-T cells and are preserved by IL-7 and IL-15. Blood. 2014;123:3750–3759. doi: 10.1182/blood-2014-01-552174. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 62.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. doi: 10.1038/s41591-018-0010-1. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 63.Gattinoni L., Klebanoff C.A., Palmer D.C., Wrzesinski C., Kerstann K.W., Yu Z., Finkelstein S.E., Theoret M.R., Rosenberg S.A., Restifo N.P. Acquisition of full effector function in vitro paradoxically impairs the in vivo antitumor efficacy of adoptively transferred CD8+ T cells. J. Clin. Investig. 2005;115:1616–1626. doi: 10.1172/JCI24480. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 64.Lopez-Cantillo G., Urueña C., Camacho B.A., Ramírez-Segura C. CAR-T Cell Performance: How to Improve Their Persistence? Front. Immunol. 2022;13:878209. doi: 10.3389/fimmu.2022.878209. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 65.Gattinoni L., Klebanoff C.A., Restifo N.P. Paths to stemness: Building the ultimate antitumour T cell. Nat. Rev. Cancer. 2012;12:671–684. doi: 10.1038/nrc3322. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 66.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 naive rather than central memory CD8+ T cells mediate superior antitumor immunity. Proc. Natl. Acad. Sci. USA. 2009;106:17469–17474. doi: 10.1073/pnas.0907448106. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 67.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. doi: 10.1073/pnas.0503726102. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 68.Meyran D., Zhu J.J., Butler J., Tantalo D., MacDonald S., Nguyen T.N., Wang M., Thio N., D’souza C., Qin V.M., et al. T(STEM)-like CAR-T cells exhibit improved persistence and tumor control compared with conventional CAR-T cells in preclinical models. Sci. Transl. Med. 2023;15:eabk1900. doi: 10.1126/scitranslmed.abk1900. [DOI] [PubMed] [Google Scholar]
- 69.Arcangeli S., Bove C., Mezzanotte C., Camisa B., Falcone L., Manfredi F., Bezzecchi E., El Khoury R., Norata R., Sanvito F., et al. CAR T cell manufacturing from naive/stem memory T lymphocytes enhances antitumor responses while curtailing cytokine release syndrome. J. Clin. Investig. 2022;132:e150807. doi: 10.1172/JCI150807. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 70.Muranski P., Borman Z.A., Kerkar S.P., Klebanoff C.A., Ji Y., Sanchez-Perez L., Sukumar M., Reger R.N., Yu Z., Kern S.J., et al. Th17 cells are long lived and retain a stem cell-like molecular signature. Immunity. 2011;35:972–985. doi: 10.1016/j.immuni.2011.09.019. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 71.Paulos C.M., Carpenito C., Plesa G., Suhoski M.M., Varela-Rohena A., Golovina T.N., Carroll R.G., Riley J.L., June C.H. The inducible costimulator (ICOS) is critical for the development of human T(H)17 cells. Sci. Transl. Med. 2010;2:55ra78. doi: 10.1126/scitranslmed.3000448. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 72.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. doi: 10.1182/blood-2013-10-535245. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 73.Bailey S.R., Nelson M.H., Majchrzak K., Bowers J.S., Wyatt M.M., Smith A.S., Neal L.R., Shirai K., Carpenito C., June C.H., et al. Human CD26(high) T cells elicit tumor immunity against multiple malignancies via enhanced migration and persistence. Nat. Commun. 2017;8:1961. doi: 10.1038/s41467-017-01867-9. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 74.Nelson M.H., Knochelmann H.M., Bailey S.R., Huff L.W., Bowers J.S., Majchrzak-Kuligowska K., Wyatt M.M., Rubinstein M.P., Mehrotra S., Nishimura M.I., et al. Identification of human CD4+ T cell populations with distinct antitumor activity. Sci. Adv. 2020;6:eaba7443. doi: 10.1126/sciadv.aba7443. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 75.Liao W., Lin J.X., Leonard W.J. Interleukin-2 at the crossroads of effector responses, tolerance, and immunotherapy. Immunity. 2013;38:13–25. doi: 10.1016/j.immuni.2013.01.004. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 76.Watanabe N., Mo F., McKenna M.K. Impact of Manufacturing Procedures on CAR T Cell Functionality. Front. Immunol. 2022;13:876339. doi: 10.3389/fimmu.2022.876339. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 77.Ding Z.C., Liu C., Cao Y., Habtetsion T., Kuczma M., Pi W., Kong H., Cacan E., Greer S.F., Cui Y., et al. IL-7 signaling imparts polyfunctionality and stemness potential to CD4+ T cells. Oncoimmunology. 2016;5:e1171445. doi: 10.1080/2162402X.2016.1171445. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 78.Sportes C., Hakim F.T., Memon S.A., Zhang H., Chua K.S., Brown M.R., Fleisher T.A., Krumlauf M.C., Babb R.R., Chow C.K., et al. Administration of rhIL-7 in humans increases in vivo TCR repertoire diversity by preferential expansion of naive T cell subsets. J. Exp. Med. 2008;205:1701–1714. doi: 10.1084/jem.20071681. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 79.Alizadeh D., Wong R.A., Yang X., Wang D., Pecoraro J.R., Kuo C.-F., Aguilar B., Qi Y., Ann D.K., Starr R., et al. IL15 Enhances CAR-T Cell Antitumor Activity by Reducing mTORC1 Activity and Preserving Their Stem Cell Memory Phenotype. Cancer Immunol. Res. 2019;7:759–772. doi: 10.1158/2326-6066.CIR-18-0466. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 80.Zhang K., Hu W., Li F., Wen C., Zhou L., Zhang L., Lian J., Liu S., Wang S., Zhang Y. IL-24 improves efficacy of CAR-T cell therapy by targeting stemness of tumor cells. Br. J. Cancer. 2024;130:1337–1347. doi: 10.1038/s41416-024-02601-1. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 81.Chen Y., Yu F., Jiang Y., Chen J., Wu K., Chen X., Lin Y., Zhang H., Li L., Zhang Y. Adoptive Transfer of Interleukin-21-stimulated Human CD8+ T Memory Stem Cells Efficiently Inhibits Tumor Growth. J. Immunother. 2018;41:274–283. doi: 10.1097/CJI.0000000000000229. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 82.Luo M., Gong W., Zhang Y., Li H., Ma D., Wu K., Gao Q., Fang Y. New insights into the stemness of adoptively transferred T cells by γc family cytokines. Cell Commun. Signal. 2023;21:347. doi: 10.1186/s12964-023-01354-3. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 83.Alvarez-Fernández C., Escribà-Garcia L., Vidal S., Sierra J., Briones J. A short CD3/CD28 costimulation combined with IL-21 enhance the generation of human memory stem T cells for adoptive immunotherapy. J. Transl. Med. 2016;14:214. doi: 10.1186/s12967-016-0973-y. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 84.Fung-Leung W.P. Phosphoinositide 3-kinase delta (PI3Kdelta) in leukocyte signaling and function. Cell Signal. 2011;23:603–608. doi: 10.1016/j.cellsig.2010.10.002. [DOI] [PubMed] [Google Scholar]
- 85.Soond D.R., Bjørgo E., Moltu K., Dale V.Q., Patton D.T., Torgersen K.M., Galleway F., Twomey B., Clark J., Gaston J.S.H., et al. PI3K p110delta regulates T-cell cytokine production during primary and secondary immune responses in mice and humans. Blood. 2010;115:2203–2213. doi: 10.1182/blood-2009-07-232330. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 86.Kim E.H., Suresh M. Role of PI3K/Akt signaling in memory CD8 T cell differentiation. Front. Immunol. 2013;4:20. doi: 10.3389/fimmu.2013.00020. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 87.Majchrzak K., Nelson K.H., Bowers J.S., Bailey S.R., Wyatt M.M., Wrangle J.M., Rubinstein M.P., Varela J.C., Li Z., Himes R.A., et al. beta-catenin and PI3Kdelta inhibition expands precursor Th17 cells with heightened stemness and antitumor activity. JCI Insight. 2017;2:e90547. doi: 10.1172/jci.insight.90547. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 88.Rangel Rivera G.O., Dwyer C.J., Knochelmann H.M., Smith A.S., Aksoy B.A., Cole A.C., Wyatt M.M., Kumaresan S., Thaxton J.E., Lesinski G.B., et al. Progressively Enhancing Stemness of Adoptively Transferred T Cells with PI3Kdelta Blockade Improves Metabolism and Antitumor Immunity. Cancer Res. 2024;84:69–83. doi: 10.1158/0008-5472.CAN-23-0801. [DOI] [PubMed] [Google Scholar]
- 89.Dwyer C.J., Arhontoulis D.C., Rangel Rivera G.O., Knochelmann H.M., Smith A.S., Wyatt M.M., Rubinstein M.P., Atkinson C., Thaxton J.E., Neskey D.M., et al. Ex vivo blockade of PI3K gamma or delta signaling enhances the antitumor potency of adoptively transferred CD8+ T cells. Eur. J. Immunol. 2020;50:1386–1399. doi: 10.1002/eji.201948455. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 90.Funk C.R., Wang S., Chen K.Z., Waller A., Sharma A., Edgar C.L., Gupta V.A., Chandrakasan S., Zoine J.T., Fedanov A., et al. PI3Kδ/γ inhibition promotes human CART cell epigenetic and metabolic reprogramming to enhance antitumor cytotoxicity. Blood. 2022;139:523–537. doi: 10.1182/blood.2021011597. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 91.Zheng W., O’hear C.E., Alli R., Basham J.H., Abdelsamed H.A., Palmer L.E., Jones L.L., Youngblood B., Geiger T.L. PI3K orchestration of the in vivo persistence of chimeric antigen receptor-modified T cells. Leukemia. 2018;32:1157–1167. doi: 10.1038/s41375-017-0008-6. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 92.Abu Eid R., Ahmad S., Lin Y., Webb M., Berrong Z., Shrimali R., Kumai T., Ananth S., Rodriguez P.C., Celis E., et al. Enhanced Therapeutic Efficacy and Memory of Tumor-Specific CD8 T Cells by Ex Vivo PI3K-delta Inhibition. Cancer Res. 2017;77:4135–4145. doi: 10.1158/0008-5472.CAN-16-1925. [DOI] [PubMed] [Google Scholar]
- 93.Bowers J.S., Majchrzak K., Nelson M.H., Aksoy B.A., Wyatt M.M., Smith A.S., Bailey S.R., Neal L.R., Hammerbacher J.E., Paulos C.M. PI3Kdelta Inhibition Enhances the Antitumor Fitness of Adoptively Transferred CD8+ T Cells. Front. Immunol. 2017;8:1221. doi: 10.3389/fimmu.2017.01221. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 94.Rao R.R., Li Q., Odunsi K., Shrikant P.A. The mTOR kinase determines effector versus memory CD8+ T cell fate by regulating the expression of transcription factors T-bet and Eomesodermin. Immunity. 2010;32:67–78. doi: 10.1016/j.immuni.2009.10.010. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 95.Zhu M., Han Y., Gu T., Wang R., Si X., Kong D., Zhao P., Wang X., Li J., Zhai X., et al. Class I HDAC inhibitors enhance antitumor efficacy and persistence of CAR-T cells by activation of the Wnt pathway. Cell Rep. 2024;43:114065. doi: 10.1016/j.celrep.2024.114065. [DOI] [PubMed] [Google Scholar]
- 96.Zhang Z., Li F., Tian Y., Cao L., Gao Q., Zhang C., Zhang K., Shen C., Ping Y., Maimela N.R., et al. Metformin Enhances the Antitumor Activity of CD8+ T Lymphocytes via the AMPK-miR-107-Eomes-PD-1 Pathway. J. Immunol. 2020;204:2575–2588. doi: 10.4049/jimmunol.1901213. [DOI] [PubMed] [Google Scholar]
- 97.Gattinoni L., Zhong X.-S., Palmer D.C., Ji Y., Hinrichs C.S., Yu Z., Wrzesinski C., Boni A., Cassard L., Garvin L.M., et al. Wnt signaling arrests effector T cell differentiation and generates CD8+ memory stem cells. Nat. Med. 2009;15:808–813. doi: 10.1038/nm.1982. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 98.Wyatt M.M., Huff L.W., Nelson M.H., Neal L.R., Medvec A.R., Rivera G.O.R., Smith A.S., Reyes A.M.R., Knochelmann H.M., Riley J.L., et al. Augmenting TCR signal strength and ICOS costimulation results in metabolically fit and therapeutically potent human CAR Th17 cells. Mol. Ther. 2023;31:2120–2131. doi: 10.1016/j.ymthe.2023.04.010. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 99.Zannikou M., Duffy J.T., Levine R.N., Seblani M., Liu Q., Presser A., Arrieta V.A., Chen C.J., Sonabend A.M., Horbinski C.M., et al. IL15 modification enables CAR T cells to act as a dual targeting agent against tumor cells and myeloid-derived suppressor cells in GBM. J. Immunother. Cancer. 2023;11:e006239. doi: 10.1136/jitc-2022-006239. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 100.Brimnes M.K., Gang A.O., Donia M., Straten P.T., Svane I.M., Hadrup S.R. Generation of autologous tumor-specific T cells for adoptive transfer based on vaccination, in vitro restimulation and CD3/CD28 dynabead-induced T cell expansion. Cancer Immunol. Immunother. 2012;61:1221–1231. doi: 10.1007/s00262-011-1199-8. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 101.Hellstrom I., Ledbetter J.A., Scholler N., Yang Y., Ye Z., Goodman G., Pullman J., Hayden-Ledbetter M., Hellström K.E. CD3-mediated activation of tumor-reactive lymphocytes from patients with advanced cancer. Proc. Natl. Acad. Sci. USA. 2001;98:6783–6788. doi: 10.1073/pnas.021557498. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 102.Ghassemi S., Durgin J.S., Nunez-Cruz S., Patel J., Leferovich J., Pinzone M., Shen F., Cummins K.D., Plesa G., Cantu V.A., et al. Rapid manufacturing of non-activated potent CAR T cells. Nat. Biomed. Eng. 2022;6:118–128. doi: 10.1038/s41551-021-00842-6. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 103.Song H.W., Prochazkova M., Shao L., Traynor R., Underwood S., Black M., Fellowes V., Shi R., Pouzolles M., Chou H.-C., et al. CAR-T cell expansion platforms yield distinct T cell differentiation states. Cytotherapy, 2024. Cytotherapy. 2024;26:757–768. doi: 10.1016/j.jcyt.2024.03.003. [DOI] [PubMed] [Google Scholar]
- 104.Labbé R.P., Vessillier S., Rafiq Q.A. Lentiviral Vectors for T Cell Engineering: Clinical Applications, Bioprocessing and Future Perspectives. Viruses. 2021;13:1528. doi: 10.3390/v13081528. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 105.Nakazawa Y., Huye L.E., Dotti G., Foster A.E., Vera J.F., Manuri P.R., June C.H., Rooney C.M., Wilson M.H. Optimization of the PiggyBac transposon system for the sustained genetic modification of human T lymphocytes. J. Immunother. 2009;32:826–836. doi: 10.1097/CJI.0b013e3181ad762b. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 106.Nakazawa Y., Huye L.E., Salsman V.S., Leen A.N., Ahmed N., Rollins L., Dotti G., Gottschalk S.M., Wilson M.H., Rooney C.M. PiggyBac-mediated cancer immunotherapy using EBV-specific cytotoxic T-cells expressing HER2-specific chimeric antigen receptor. Mol. Ther. 2011;19:2133–2143. doi: 10.1038/mt.2011.131. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 107.Morita D., Nishio N., Saito S., Tanaka M., Kawashima N., Okuno Y., Suzuki S., Matsuda K., Maeda Y., Wilson M.H., et al. Enhanced Expression of Anti-CD19 Chimeric Antigen Receptor in piggyBac Transposon-Engineered T Cells. Mol. Ther. Methods Clin. Dev. 2018;8:131–140. doi: 10.1016/j.omtm.2017.12.003. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 108.Zhang Y., Zhang Z., Ding Y., Fang Y., Wang P., Chu W., Jin Z., Yang X., Wang J., Lou J., et al. Phase I clinical trial of EGFR-specific CAR-T cells generated by the piggyBac transposon system in advanced relapsed/refractory non-small cell lung cancer patients. J. Cancer Res. Clin. Oncol. 2021;147:3725–3734. doi: 10.1007/s00432-021-03613-7. [DOI] [PubMed] [Google Scholar]
- 109.Ayala Ceja M., Khericha M., Harris C.M., Puig-Saus C., Chen Y.Y. CAR-T cell manufacturing: Major process parameters and next-generation strategies. J. Exp. Med. 2024;221:e20230903. doi: 10.1084/jem.20230903. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 110.Kim M.Y., Jayasinghe R., Devenport J.M., Ritchey J.K., Rettig M.P., O’neal J., Staser K.W., Kennerly K.M., Carter A.J., Gao F., et al. A long-acting interleukin-7, rhIL-7-hyFc, enhances CAR T cell expansion, persistence, and anti-tumor activity. Nat. Commun. 2022;13:3296. doi: 10.1038/s41467-022-30860-0. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 111.Klebanoff C.A., Finkelstein S.E., Surman D.R., Lichtman M.K., Gattinoni L., Theoret M.R., Grewal N., Spiess P.J., Antony P.A., Palmer D.C., et al. IL-15 enhances the in vivo antitumor activity of tumor-reactive CD8+ T cells. Proc. Natl. Acad. Sci. USA. 2004;101:1969–1974. doi: 10.1073/pnas.0307298101. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 112.Pilipow K., Roberto A., Roederer M., Waldmann T.A., Mavilio D., Lugli E. IL15 and T-cell Stemness in T-cell-Based Cancer Immunotherapy. Cancer Res. 2015;75:5187–5193. doi: 10.1158/0008-5472.CAN-15-1498. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 113.Conlon K.C., Lugli E., Welles H.C., Rosenberg S.A., Fojo A.T., Morris J.C., Fleisher T.A., Dubois S.P., Perera L.P., Stewart D.M., et al. Redistribution, hyperproliferation, activation of natural killer cells and CD8 T cells, and cytokine production during first-in-human clinical trial of recombinant human interleukin-15 in patients with cancer. J. Clin. Oncol. 2015;33:74–82. doi: 10.1200/JCO.2014.57.3329. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 114.Nguyen R., Doubrovina E., Mousset C.M., Jin B.Y., Okada R., Zhang X., Clavel A., Reyes-Gonzalez J.M., Dyomin V., Diaz L., et al. Cooperative Armoring of CAR and TCR T Cells by T Cell-Restricted IL15 and IL21 Universally Enhances Solid Tumor Efficacy. Clin. Cancer Res. 2024;30:1555–1566. doi: 10.1158/1078-0432.CCR-23-1872. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 115.Hurton L.V., Singh H., Najjar A.M., Switzer K.C., Mi T., Maiti S., Olivares S., Rabinovich B., Huls H., Forget M.-A., et al. Tethered IL-15 augments antitumor activity and promotes a stem-cell memory subset in tumor-specific T cells. Proc. Natl. Acad. Sci. USA. 2016;113:E7788–E7797. doi: 10.1073/pnas.1610544113. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 116.Atkins M.B., Sparano J., I Fisher R., Weiss G.R., A Margolin K., I Fink K., Rubinstein L., Louie A., Mier J.W., Gucalp R. Randomized phase II trial of high-dose interleukin-2 either alone or in combination with interferon alfa-2b in advanced renal cell carcinoma. J. Clin. Oncol. 1993;11:661–670. doi: 10.1200/JCO.1993.11.4.661. [DOI] [PubMed] [Google Scholar]
- 117.Atkins M.B., Lotze M.T., Dutcher J.P., Fisher R.I., Weiss G., Margolin K., Abrams J., Sznol M., Parkinson D., Hawkins M., et al. High-dose recombinant interleukin 2 therapy for patients with metastatic melanoma: Analysis of 270 patients treated between 1985 and 1993. J. Clin. Oncol. 1999;17:2105–2116. doi: 10.1200/JCO.1999.17.7.2105. [DOI] [PubMed] [Google Scholar]
- 118.Fyfe G., Fisher R.I., Rosenberg S.A., Sznol M., Parkinson D.R., Louie A.C. Results of treatment of 255 patients with metastatic renal cell carcinoma who received high-dose recombinant interleukin-2 therapy. J. Clin. Oncol. 1995;13:688–696. doi: 10.1200/JCO.1995.13.3.688. [DOI] [PubMed] [Google Scholar]
- 119.Zhang Q., Hresko M.E., Picton L.K., Su L., Hollander M.J., Nunez-Cruz S., Zhang Z., Assenmacher C.A., Sockolosky J.T., Garcia K.C., et al. A human orthogonal IL-2 and IL-2Rbeta system enhances CAR T cell expansion and antitumor activity in a murine model of leukemia. Sci. Transl. Med. 2021;13:eabg6986. doi: 10.1126/scitranslmed.abg6986. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 120.Koneru M., Purdon T.J., Spriggs D., Koneru S., Brentjens R.J. IL-12 secreting tumor-targeted chimeric antigen receptor T cells eradicate ovarian tumors in vivo. Oncoimmunology. 2015;4:e994446. doi: 10.4161/2162402X.2014.994446. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 121.Kueberuwa G., Kalaitsidou M., Cheadle E., Hawkins R.E., Gilham D.E. CD19 CAR T Cells Expressing IL-12 Eradicate Lymphoma in Fully Lymphoreplete Mice through Induction of Host Immunity. Mol. Ther. Oncolytics. 2018;8:41–51. doi: 10.1016/j.omto.2017.12.003. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 122.Leonard J.P., Sherman M.L., Fisher G.L., Buchanan L.J., Larsen G., Atkins M.B., A Sosman J., Dutcher J.P., Vogelzang N.J., Ryan J.L. Effects of single-dose interleukin-12 exposure on interleukin-12-associated toxicity and interferon-gamma production. Blood. 1997;90:2541–2548. [PubMed] [Google Scholar]
- 123.Yeku O.O., Purdon T.J., Koneru M., Spriggs D., Brentjens R.J. Armored CAR T cells enhance antitumor efficacy and overcome the tumor microenvironment. Sci. Rep. 2017;7:10541. doi: 10.1038/s41598-017-10940-8. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 124.Agliardi G., Liuzzi A.R., Hotblack A., De Feo D., Núñez N., Stowe C.L., Friebel E., Nannini F., Rindlisbacher L., Roberts T.A., et al. Intratumoral IL-12 delivery empowers CAR-T cell immunotherapy in a pre-clinical model of glioblastoma. Nat. Commun. 2021;12:444. doi: 10.1038/s41467-020-20599-x. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 125.Anwer K., Barnes M.N., Fewell J., Lewis D.H., Alvarez R.D. Phase-I clinical trial of IL-12 plasmid/lipopolymer complexes for the treatment of recurrent ovarian cancer. Gene Ther. 2010;17:360–369. doi: 10.1038/gt.2009.159. [DOI] [PubMed] [Google Scholar]
- 126.Ma X., Shou P., Smith C., Chen Y., Du H., Sun C., Kren N.P., Michaud D., Ahn S., Vincent B., et al. Interleukin-23 engineering improves CAR T cell function in solid tumors. Nat. Biotechnol. 2020;38:448–459. doi: 10.1038/s41587-019-0398-2. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 127.Svoboda J., Gerson J.N., Landsburg D.J., Chong E.A., Barta S.K., Nasta S.D., Ruella M., Hexner E.O., Marshall A., Leskowitz R., et al. Interleukin-18 Secreting Autologous Anti-CD19 CAR T-Cells (huCART19-IL18) in Patients with Non-Hodgkin Lymphomas Relapsed or Refractory to Prior CAR T-Cell Therapy. Blood. 2022;140((Suppl. S1)):4612–4614. doi: 10.1182/blood-2022-162393. [DOI] [Google Scholar]
- 128.Chmielewski M., Abken H. CAR T Cells Releasing IL-18 Convert to T-Bet(high) FoxO1(low) Effectors that Exhibit Augmented Activity against Advanced Solid Tumors. Cell Rep. 2017;21:3205–3219. doi: 10.1016/j.celrep.2017.11.063. [DOI] [PubMed] [Google Scholar]
- 129.Nagarsheth N., Wicha M.S., Zou W. Chemokines in the cancer microenvironment and their relevance in cancer immunotherapy. Nat. Rev. Immunol. 2017;17:559–572. doi: 10.1038/nri.2017.49. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 130.Foeng J., Comerford I., McColl S.R. Harnessing the chemokine system to home CAR-T cells into solid tumors. Cell Rep. Med. 2022;3:100543. doi: 10.1016/j.xcrm.2022.100543. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 131.Kankeu Fonkoua L.A., Sirpilla O., Sakemura R., Siegler E.L., Kenderian S.S. CAR T cell therapy and the tumor microenvironment: Current challenges and opportunities. Mol. Ther. Oncolytics. 2022;25:69–77. doi: 10.1016/j.omto.2022.03.009. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 132.Harlin H., Meng Y., Peterson A.C., Zha Y., Tretiakova M., Slingluff C., McKee M., Gajewski T.F. Chemokine expression in melanoma metastases associated with CD8+ T-cell recruitment. Cancer Res. 2009;69:3077–3085. doi: 10.1158/0008-5472.CAN-08-2281. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 133.Zhang L., Conejo-Garcia J.R., Katsaros D., Gimotty P.A., Massobrio M., Regnani G., Makrigiannakis A., Gray H., Schlienger K., Liebman M.N., et al. Intratumoral T cells, recurrence, and survival in epithelial ovarian cancer. N. Engl. J. Med. 2003;348:203–213. doi: 10.1056/NEJMoa020177. [DOI] [PubMed] [Google Scholar]
- 134.Lesch S., Blumenberg V., Stoiber S., Gottschlich A., Ogonek J., Cadilha B.L., Dantes Z., Rataj F., Dorman K., Lutz J., et al. T cells armed with C-X-C chemokine receptor type 6 enhance adoptive cell therapy for pancreatic tumours. Nat. Biomed. Eng. 2021;5:1246–1260. doi: 10.1038/s41551-021-00737-6. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 135.Whilding L.M., Halim L., Draper B., Parente-Pereira A.C., Zabinski T., Davies D.M., Maher J. CAR T-Cells Targeting the Integrin alphavbeta6 and Co-Expressing the Chemokine Receptor CXCR2 Demonstrate Enhanced Homing and Efficacy against Several Solid Malignancies. Cancers. 2019;11:674. doi: 10.3390/cancers11050674. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 136.Shah N.N., Johnson B.D., Schneider D., Zhu F., Szabo A., Keever-Taylor C.A., Krueger W., Worden A.A., Kadan M.J., Yim S., et al. Bispecific anti-CD20, anti-CD19 CAR T cells for relapsed B cell malignancies: A phase 1 dose escalation and expansion trial. Nat. Med. 2020;26:1569–1575. doi: 10.1038/s41591-020-1081-3. [DOI] [PubMed] [Google Scholar]
- 137.Ying Z., Huang X.F., Xiang X., Liu Y., Kang X., Song Y., Guo X., Liu H., Ding N., Zhang T., et al. A safe and potent anti-CD19 CAR T cell therapy. Nat. Med. 2019;25:947–953. doi: 10.1038/s41591-019-0421-7. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 138.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. doi: 10.1182/blood-2011-04-348540. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 139.Kalos M., Levine B.L., Porter D.L., Katz S., 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. doi: 10.1126/scitranslmed.3002842. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 140.Haas A.R., Tanyi J.L., O’hara M.H., Gladney W.L., Lacey S.F., Torigian D.A., Soulen M.C., Tian L., McGarvey M., Nelson A.M., et al. Phase I Study of Lentiviral-Transduced Chimeric Antigen Receptor-Modified T Cells Recognizing Mesothelin in Advanced Solid Cancers. Mol. Ther. 2019;27:1919–1929. doi: 10.1016/j.ymthe.2019.07.015. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 141.Knochelmann H.M., Dwyer C.J., Smith A.S., Bowers J.S., Wyatt M.M., Nelson M.H., Rivera G.O.R., Horton J.D., Krieg C., Armeson K., et al. IL6 Fuels Durable Memory for Th17 Cell-Mediated Responses to Tumors. Cancer Res. 2020;80:3920–3932. doi: 10.1158/0008-5472.CAN-19-3685. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 142.Dickinson M.J., Barba P., Jäger U., Shah N.N., Blaise D., Briones J., Shune L., Boissel N., Bondanza A., Mariconti L., et al. A Novel Autologous CAR-T Therapy, YTB323, with Preserved T-cell Stemness Shows Enhanced CAR T-cell Efficacy in Preclinical and Early Clinical Development. Cancer Discov. 2023;13:1982–1997. doi: 10.1158/2159-8290.CD-22-1276. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 143.Ikegawa S., Sperling A.S., Ansuinelli M., Nikiforow S., Quinn D., Bu D., Mataraza J., Pearson D., Rispoli L., Credi M.A., et al. T-Charge™ Manufacturing of the Anti-BCMA CAR-T, Durcabtagene Autoleucel (PHE885), Promotes Expansion and Persistence of CAR-T Cells with High TCR Repertoire Diversity. Blood. 2023;142((Suppl. S1)):3469. doi: 10.1182/blood-2023-177721. [DOI] [Google Scholar]
- 144.Klaver Y., van Steenbergen S.C.L., Sleijfer S., Debets R., Lamers C.H.J. T Cell Maturation Stage Prior to and During GMP Processing Informs on CAR T Cell Expansion in Patients. Front. Immunol. 2016;7:648. doi: 10.3389/fimmu.2016.00648. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 145.Wrzesinski C., Paulos C.M., Kaiser A., Muranski P., Palmer D.C., Gattinoni L., Yu Z., Rosenberg S.A., Restifo N.P. Increased intensity lymphodepletion enhances tumor treatment efficacy of adoptively transferred tumor-specific T cells. J. Immunother. 2010;33:1–7. doi: 10.1097/CJI.0b013e3181b88ffc. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 146.Gattinoni L., Finkelstein S.E., Klebanoff C.A., Antony P.A., Palmer D.C., Spiess P.J., Hwang L.N., Yu Z., Wrzesinski C., Heimann D.M., et al. Removal of homeostatic cytokine sinks by lymphodepletion enhances the efficacy of adoptively transferred tumor-specific CD8+ T cells. J. Exp. Med. 2005;202:907–912. doi: 10.1084/jem.20050732. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 147.Paulos C.M., Wrzesinski C., Kaiser A., Hinrichs C.S., Chieppa M., Cassard L., Palmer D.C., Boni A., Muranski P., Yu Z., et al. Microbial translocation augments the function of adoptively transferred self/tumor-specific CD8+ T cells via TLR4 signaling. J. Clin. Investig. 2007;117:2197–2204. doi: 10.1172/JCI32205. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 148.Antony P.A., Piccirillo C.A., Akpinarli A., Finkelstein S.E., Speiss P.J., Surman D.R., Palmer D.C., Chan C.-C., Klebanoff C.A., Overwijk W.W., et al. CD8+ T cell immunity against a tumor/self-antigen is augmented by CD4+ T helper cells and hindered by naturally occurring T regulatory cells. J. Immunol. 2005;174:2591–2601. doi: 10.4049/jimmunol.174.5.2591. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 149.Klebanoff C.A., Khong H.T., Antony P.A., Palmer D.C., Restifo N.P. Sinks, suppressors and antigen presenters: How lymphodepletion enhances T cell-mediated tumor immunotherapy. Trends Immunol. 2005;26:111–117. doi: 10.1016/j.it.2004.12.003. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 150.Citrin D.E. Recent Developments in Radiotherapy. N. Engl. J. Med. 2017;377:2200–2201. doi: 10.1056/NEJMra1608986. [DOI] [PubMed] [Google Scholar]
- 151.Lhuillier C., Rudqvist N.-P., Elemento O., Formenti S.C., Demaria S. Radiation therapy and anti-tumor immunity: Exposing immunogenic mutations to the immune system. Genome Med. 2019;11:40. doi: 10.1186/s13073-019-0653-7. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 152.Fabrizio V.A., Boelens J.J., Mauguen A., Baggott C., Prabhu S., Egeler E., Mavroukakis S., Pacenta H.L., Phillips C.L., Rossoff J., et al. Optimal fludarabine lymphodepletion is associated with improved outcomes after CAR T-cell therapy. Blood Adv. 2022;6:1961–1968. doi: 10.1182/bloodadvances.2021006418. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 153.Hirayama A.V., Gauthier J., Hay K.A., Voutsinas J.M., Wu Q., Gooley T., Li D., Cherian S., Chen X., Pender B.S., et al. The response to lymphodepletion impacts PFS in patients with aggressive non-Hodgkin lymphoma treated with CD19 CAR T cells. Blood. 2019;133:1876–1887. doi: 10.1182/blood-2018-11-887067. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 154.Lickefett B., Chu L., Ortiz-Maldonado V., Warmuth L., Barba P., Doglio M., Henderson D., Hudecek M., Kremer A., Markman J., et al. Lymphodepletion—an essential but undervalued part of the chimeric antigen receptor T-cell therapy cycle. Front. Immunol. 2023;14:1303935. doi: 10.3389/fimmu.2023.1303935. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 155.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. doi: 10.1172/JCI85309. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 156.Wittling M.C., Knochelmann H.M., Wyatt M.M., Rangel Rivera G.O., Cole A.C., Lesinski G.B., Paulos C.M. Distinct host preconditioning regimens differentially impact the antitumor potency of adoptively transferred Th17 cells. J. Immunother. Cancer. 2024;12:e008715. doi: 10.1136/jitc-2023-008715. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 157.Suryadevara C.M., Desai R., Abel M.L., Riccione K.A., Batich K.A., Shen S.H., Chongsathidkiet P., Gedeon P.C., Elsamadicy A.A., Snyder D.J., et al. Temozolomide lymphodepletion enhances CAR abundance and correlates with antitumor efficacy against established glioblastoma. Oncoimmunology. 2018;7:e1434464. doi: 10.1080/2162402X.2018.1434464. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 158.Wang Z., Chen M., Zhang Y., Liu Y., Yang Q., Nie J., Shen L., Jiang P., He J., Ye X., et al. Phase I study of CRISPR-engineered CAR-T cells with PD-1 inactivation in treating mesothelin-positive solid tumors. J. Clin. Oncol. 2020;38((Suppl. S15)):3038. doi: 10.1200/JCO.2020.38.15_suppl.3038. [DOI] [Google Scholar]
- 159.Hegde M., Navai S., DeRenzo C., Joseph S.K., Sanber K., Wu M., Gad A.Z., Janeway K.A., Campbell M., Mullikin D., et al. Autologous HER2-specific CAR T cells after lymphodepletion for advanced sarcoma: A phase 1 trial. Nat. Cancer. 2024;5:880–894. doi: 10.1038/s43018-024-00749-6. [DOI] [PubMed] [Google Scholar]
- 160.Dorff T.B., Blanchard M.S., Adkins L.N., Luebbert L., Leggett N., Shishido S.N., Macias A., Del Real M.M., Dhapola G., Egelston C., et al. PSCA-CAR T cell therapy in metastatic castration-resistant prostate cancer: A phase 1 trial. Nat. Med. 2024;30:1636–1644. doi: 10.1038/s41591-024-02979-8. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 161.Wang A.X., Ong X.J., D’souza C., Neeson P.J., Zhu J.J. Combining chemotherapy with CAR-T cell therapy in treating solid tumors. Front. Immunol. 2023;14:1140541. doi: 10.3389/fimmu.2023.1140541. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 162.Goff S.L., Dudley M.E., Citrin D.E., Somerville R.P., Wunderlich J.R., Danforth D.N., Zlott D.A., Yang J.C., Sherry R.M., Kammula U.S., et al. Randomized, Prospective Evaluation Comparing Intensity of Lymphodepletion Before Adoptive Transfer of Tumor-Infiltrating Lymphocytes for Patients with Metastatic Melanoma. J. Clin. Oncol. 2016;34:2389–2397. doi: 10.1200/JCO.2016.66.7220. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 163.Dudley M.E., Yang J.C., Sherry R., Hughes M.S., Royal R., Kammula U., Robbins P.F., Huang J., Citrin D.E., Leitman S.F., et al. Adoptive cell therapy for patients with metastatic melanoma: Evaluation of intensive myeloablative chemoradiation preparative regimens. J. Clin. Oncol. 2008;26:5233–5239. doi: 10.1200/JCO.2008.16.5449. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 164.Brog R.A., Ferry S.L., Schiebout C.T., Messier C.M., Cook W.J., Abdullah L., Zou J., Kumar P., Sentman C.L., Frost H.R., et al. Superkine IL-2 and IL-33 Armored CAR T Cells Reshape the Tumor Microenvironment and Reduce Growth of Multiple Solid Tumors. Cancer Immunol. Res. 2022;10:962–977. doi: 10.1158/2326-6066.CIR-21-0536. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 165.Luo H., Su J., Sun R., Sun Y., Wang Y., Dong Y., Shi B., Jiang H., Li Z. Coexpression of IL7 and CCL21 Increases Efficacy of CAR-T Cells in Solid Tumors without Requiring Preconditioned Lymphodepletion. Clin. Cancer Res. 2020;26:5494–5505. doi: 10.1158/1078-0432.CCR-20-0777. [DOI] [PubMed] [Google Scholar]
- 166.Tennant M.D., New C., Ferreira L.M., O’neil R.T. Efficient T cell adoptive transfer in lymphoreplete hosts mediated by transient activation of Stat5 signaling. Mol. Ther. 2023;31:2591–2599. doi: 10.1016/j.ymthe.2023.07.015. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 167.Choi B.D., Gerstner E.R., Frigault M.J., Leick M.B., Mount C.W., Balaj L., Nikiforow S., Carter B.S., Curry W.T., Gallagher K., et al. Intraventricular CARv3-TEAM-E T Cells in Recurrent Glioblastoma. N. Engl. J. Med. 2024;390:1290–1298. doi: 10.1056/NEJMoa2314390. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 168.Brown C.E., Alizadeh D., Starr R., Weng L., Wagner J.R., Naranjo A., Ostberg J.R., Blanchard M.S., Kilpatrick J., Simpson J., et al. Regression of Glioblastoma after Chimeric Antigen Receptor T-Cell Therapy. N. Engl. J. Med. 2016;375:2561–2569. doi: 10.1056/NEJMoa1610497. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 169.Bagley S.J., Logun M., Fraietta J.A., Wang X., Desai A.S., Bagley L.J., Nabavizadeh A., Jarocha D., Martins R., Maloney E., et al. Intrathecal bivalent CAR T cells targeting EGFR and IL13Ralpha2 in recurrent glioblastoma: Phase 1 trial interim results. Nat. Med. 2024;30:1320–1329. doi: 10.1038/s41591-024-02893-z. [DOI] [PubMed] [Google Scholar]
- 170.Brown C.E., Hibbard J.C., Alizadeh D., Blanchard M.S., Natri H.M., Wang D., Ostberg J.R., Aguilar B., Wagner J.R., Paul J.A., et al. Locoregional delivery of IL-13Ralpha2-targeting CAR-T cells in recurrent high-grade glioma: A phase 1 trial. Nat. Med. 2024;30:1001–1012. doi: 10.1038/s41591-024-02875-1. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 171.Bagley S.J., Binder Z.A., Lamrani L., Marinari E., Desai A.S., Nasrallah M.P., Maloney E., Brem S., Lustig R.A., Kurtz G., et al. Repeated peripheral infusions of anti-EGFRvIII CAR T cells in combination with pembrolizumab show no efficacy in glioblastoma: A phase 1 trial. Nat. Cancer. 2024;5:517–531. doi: 10.1038/s43018-023-00709-6. [DOI] [PubMed] [Google Scholar]
- 172.Goff S.L., Morgan R.A., Yang J.C., Sherry R.M., Robbins P.F., Restifo N.P., Feldman S.A., Lu Y.-C., Lu L., Zheng Z., et al. Pilot Trial of Adoptive Transfer of Chimeric Antigen Receptor-transduced T Cells Targeting EGFRvIII in Patients with Glioblastoma. J. Immunother. 2019;42:126–135. doi: 10.1097/CJI.0000000000000260. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 173.Huang Z., Dewanjee S., Chakraborty P., Jha N.K., Dey A., Gangopadhyay M., Chen X.-Y., Wang J., Jha S.K. CAR T cells: Engineered immune cells to treat brain cancers and beyond. Mol. Cancer. 2023;22:22. doi: 10.1186/s12943-022-01712-8. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 174.Aggarwal V., Workman C.J., Vignali D.A.A. LAG-3 as the third checkpoint inhibitor. Nat. Immunol. 2023;24:1415–1422. doi: 10.1038/s41590-023-01569-z. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 175.Postow M.A., Callahan M.K., Wolchok J.D. Immune Checkpoint Blockade in Cancer Therapy. J. Clin. Oncol. 2015;33:1974–1982. doi: 10.1200/JCO.2014.59.4358. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 176.Wei S.C., Duffy C.R., Allison J.P. Fundamental Mechanisms of Immune Checkpoint Blockade Therapy. Cancer Discov. 2018;8:1069–1086. doi: 10.1158/2159-8290.CD-18-0367. [DOI] [PubMed] [Google Scholar]
- 177.Chong E.A., Melenhorst J.J., Lacey S.F., Ambrose D.E., Gonzalez V., Levine B.L., June C.H., Schuster S.J. PD-1 blockade modulates chimeric antigen receptor (CAR)-modified T cells: Refueling the CAR. Blood. 2017;129:1039–1041. doi: 10.1182/blood-2016-09-738245. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 178.Grosser R., Cherkassky L., Chintala N., Adusumilli P.S. Combination Immunotherapy with CAR T Cells and Checkpoint Blockade for the Treatment of Solid Tumors. Cancer Cell. 2019;36:471–482. doi: 10.1016/j.ccell.2019.09.006. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 179.Adusumilli P.S., Zauderer M.G., Rivière I., Solomon S.B., Rusch V.W., O’Cearbhaill R.E., Zhu A., Cheema W., Chintala N.K., Halton E., et al. A Phase I Trial of Regional Mesothelin-Targeted CAR T-cell Therapy in Patients with Malignant Pleural Disease, in Combination with the Anti-PD-1 Agent Pembrolizumab. Cancer Discov. 2021;11:2748–2763. doi: 10.1158/2159-8290.CD-21-0407. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 180.Maude S.L., E Hucks G., Seif A.E., Talekar M.K., Teachey D.T., Baniewicz D., Callahan C., Gonzalez V., Nazimuddin F., Gupta M., et al. The effect of pembrolizumab in combination with CD19-targeted chimeric antigen receptor (CAR) T cells in relapsed acute lymphoblastic leukemia (ALL) J. Clin. Oncol. 2017;35((Suppl. S15)):103. doi: 10.1200/JCO.2017.35.15_suppl.103. [DOI] [Google Scholar]
- 181.Maude S.L., E Hucks G., Seif A.E., Talekar M.K., Teachey D.T., Baniewicz D., Callahan C., Gonzalez V., Nazimuddin F., Gupta M., et al. Characterization of anti-CD19 chimeric antigen receptor (CAR) T cell-mediated tumor microenvironment immune gene profile in a multicenter trial (ZUMA-1) with axicabtagene ciloleucel (axi-cel, KTE-C19) J. Clin. Oncol. 2017;35((Suppl. S15)):3025. [Google Scholar]
- 182.Chong E.A., Svoboda J., Nasta S.D., Landsburg D.J., Winchell N., Napier E., Mato A.R., Melenhorst J.J., Ruella M., Lacey S.F., et al. Sequential Anti-CD19 Directed Chimeric Antigen Receptor Modified T-Cell Therapy (CART19) and PD-1 Blockade with Pembrolizumab in Patients with Relapsed or Refractory B-Cell Non-Hodgkin Lymphomas. Blood. 2018;132:4198. doi: 10.1182/blood-2018-99-119502. [DOI] [Google Scholar]
- 183.Melief C.J.M., Welters M.J.P., Vergote I., Kroep J.R., Kenter G.G., Ottevanger P.B., Tjalma W.A.A., Denys H., van Poelgeest M.I.E., Nijman H.W., et al. Strong vaccine responses during chemotherapy are associated with prolonged cancer survival. Sci. Transl. Med. 2020;12:eaaz8235. doi: 10.1126/scitranslmed.aaz8235. [DOI] [PubMed] [Google Scholar]
- 184.Bocchia M., Defina M., Aprile L., Ippoliti M., Crupi R., Rondoni M., Gozzetti A., Lauria F. Complete molecular response in CML after p210 BCR-ABL1-derived peptide vaccination. Nat. Rev. Clin. Oncol. 2010;7:600–603. doi: 10.1038/nrclinonc.2010.141. [DOI] [PubMed] [Google Scholar]
- 185.Koster B.D., Santegoets S.J.A.M., Harting J., Baars A., van Ham S.A., Scheper R.J., Hooijberg E., de Gruijl T.D., van den Eertwegh A.J.M. Autologous tumor cell vaccination combined with systemic CpG-B and IFN-alpha promotes immune activation and induces clinical responses in patients with metastatic renal cell carcinoma: A phase II trial. Cancer Immunol. Immunother. 2019;68:1025–1035. doi: 10.1007/s00262-019-02320-0. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 186.Sahin U., Oehm P., Derhovanessian E., Jabulowsky R.A., Vormehr M., Gold M., Maurus D., Schwarck-Kokarakis D., Kuhn A.N., Omokoko T., et al. An RNA vaccine drives immunity in checkpoint-inhibitor-treated melanoma. Nature. 2020;585:107–112. doi: 10.1038/s41586-020-2537-9. [DOI] [PubMed] [Google Scholar]
- 187.Saxena M., van der Burg S.H., Melief C.J.M., Bhardwaj N. Therapeutic cancer vaccines. Nat. Rev. Cancer. 2021;21:360–378. doi: 10.1038/s41568-021-00346-0. [DOI] [PubMed] [Google Scholar]
- 188.Reinhard K., Rengstl B., Oehm P., Michel K., Billmeier A., Hayduk N., Klein O., Kuna K., Ouchan Y., Wöll S., et al. An RNA vaccine drives expansion and efficacy of claudin-CAR-T cells against solid tumors. Science. 2020;367:446–453. doi: 10.1126/science.aay5967. [DOI] [PubMed] [Google Scholar]
- 189.Mackensen A., Haanen J.B., Koenecke C., Alsdorf W., Wagner-Drouet E., Borchmann P., Heudobler D., Ferstl B., Klobuch S., Bokemeyer C., et al. CLDN6-specific CAR-T cells plus amplifying RNA vaccine in relapsed or refractory solid tumors: The phase 1 BNT211-01 trial. Nat. Med. 2023;29:2844–2853. doi: 10.1038/s41591-023-02612-0. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 190.Ma L., Dichwalkar T., Chang J.Y.H., Cossette B., Garafola D., Zhang A.Q., Fichter M., Wang C., Liang S., Silva M., et al. Enhanced CAR-T cell activity against solid tumors by vaccine boosting through the chimeric receptor. Science. 2019;365:162–168. doi: 10.1126/science.aav8692. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 191.Akahori Y., Wang L., Yoneyama M., Seo N., Okumura S., Miyahara Y., Amaishi Y., Okamoto S., Mineno J., Ikeda H., et al. Antitumor activity of CAR-T cells targeting the intracellular oncoprotein WT1 can be enhanced by vaccination. Blood. 2018;132:1134–1145. doi: 10.1182/blood-2017-08-802926. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 192.Ansah E.O., Baah A., Agyenim E.B. Vaccine Boosting CAR-T Cell Therapy: Current and Future Strategies. Adv. Cell Gene Ther. 2023;2023:8030440. doi: 10.1155/2023/8030440. [DOI] [Google Scholar]
- 193.Redeker A., Arens R. Improving Adoptive T Cell Therapy: The Particular Role of T Cell Costimulation, Cytokines, and Post-Transfer Vaccination. Front. Immunol. 2016;7:345. doi: 10.3389/fimmu.2016.00345. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 194.Wei J., Han X., Bo J., Han W. Target selection for CAR-T therapy. J. Hematol. Oncol. 2019;12:62. doi: 10.1186/s13045-019-0758-x. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 195.Sterner R.C., Sterner R.M. CAR-T cell therapy: Current limitations and potential strategies. Blood Cancer J. 2021;11:69. doi: 10.1038/s41408-021-00459-7. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 196.White L.G., Goy H.E., Rose A.J., McLellan A.D. Controlling Cell Trafficking: Addressing Failures in CAR T and NK Cell Therapy of Solid Tumours. Cancers. 2022;14:978. doi: 10.3390/cancers14040978. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 197.Martinez M., Moon E.K. CAR T Cells for Solid Tumors: New Strategies for Finding, Infiltrating, and Surviving in the Tumor Microenvironment. Front. Immunol. 2019;10:128. doi: 10.3389/fimmu.2019.00128. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 198.Akbari P., Katsarou A., Daghighian R., van Mil L.W., Huijbers E.J., Griffioen A.W., van Beijnum J.R. Directing CAR T cells towards the tumor vasculature for the treatment of solid tumors. Biochim. Biophys. Acta Rev. Cancer. 2022;1877:188701. doi: 10.1016/j.bbcan.2022.188701. [DOI] [PubMed] [Google Scholar]