Skip to main content
The Journal of Clinical Investigation logoLink to The Journal of Clinical Investigation
. 2021 Jan 19;131(2):e142030. doi: 10.1172/JCI142030

Insight into next-generation CAR therapeutics: designing CAR T cells to improve clinical outcomes

Emiliano Roselli 1, Rawan Faramand 2, Marco L Davila 1,2,3,4,
PMCID: PMC7810492  PMID: 33463538

Abstract

Chimeric antigen receptor (CAR) T cell therapy has shown considerable promise for hematologic malignancies, leading to the US Food and Drug Administration approval of two CAR T cell–based therapies for the treatment of B cell acute lymphoblastic leukemia and large B cell lymphoma. Despite success in hematologic malignancies, the treatment landscape of CAR T cell therapy for solid tumors has been limited. There are unique challenges in the development of novel CAR T cell therapies to improve both safety and efficacy. Improved understanding of the immunosuppressive tumor microenvironment and resistance mechanisms has led to encouraging approaches to mitigating these obstacles. This Review will characterize challenges with current CAR T designs for hematologic malignancies and solid tumors and emphasize preclinical and clinical strategies to overcome them with novel CAR T cell therapies.

Introduction

In the last decade, the concept of harnessing the intrinsic and complex ability of the immune system to fight cancer has multiplied, with a pronounced expansion in the field of immuno-oncology demonstrating durable clinical responses in patients with solid tumors and hematologic malignancies (1). Adoptive T cell therapy (ACT) is an example of an immunotherapy employed in the clinic, a form of passive immunotherapy based on the infusion of lymphocytes to generate an antitumor response (2). Chimeric antigen receptors (CARs) are synthetic molecules that harness the antigen-recognition ability of antibodies and the effector functions of immune cells, typically T cells. T cells are genetically engineered to express CARs, which are composed of an extracellular antigen-recognition domain derived from an antibody, a hinge and transmembrane domain, and intracellular motifs that activate and augment cell function upon antigen engagement (Figure 1 and ref. 3).

Figure 1. Structure and evolution of CARs.

Figure 1

CARs are composed of an antigen-recognition domain (mainly scFv of monoclonal antibodies) fused to a hinge/transmembrane domain and intracellular signaling domains able to transduce activation signals to CAR T cells. Modification in the intracellular domains gave rise to a different generation of CAR T cells from the first generation, which included a CD3ζ motif, to the second and third generations, which included one or two costimulatory domains, respectively. VH, variable heavy chain; VL, variable light chain.

Within this field, CD19-targeted CAR T cells for the treatment of B cell malignancies were the first cell-based therapeutic approved by the US FDA in 2017 (4, 5). Despite the successes of CAR T cell therapy for B cell malignancies, immune-mediated toxicities can lead to morbidity and mortality, limiting the widespread use of this therapy. Moreover, durable responses are observed in less than half of CAR T cell–treated patients, underscoring the importance of developing next-generation CARs that are safer and more effective (68). CAR T cell therapy for solid tumors has not yet translated into clinical practice due to challenges such as tumor antigen heterogeneity, difficulty trafficking and infiltrating into the tumor sites, and an immunosuppressive tumor microenvironment (TME).

Evolution in CAR design

CAR design evolved into different generations defined by modifications of the endodomain of the receptor. First-generation CAR T cells were composed of an antigen-recognition extracellular domain joined to a single intracellular motif encoding for the cytoplasmic T cell glycoprotein CD3ζ, mimicking normal activation through the T cell receptor (TCR) (Figure 1). This initial design was able to trigger a cellular response against its target antigen (9, 10), but the modified T cells displayed an inability to persist in patients, with limited clinical benefits (11). Second-generation CAR T cells added an additional intracellular motif composed of the signaling domain of costimulatory receptors such as CD28 (12) and 4-1BB/CD137 (13) to the CD3ζ-activating domain; these modifications rendered CAR T cells with enhanced effector functions and increased persistence in vivo compared with their first-generation counterparts (14). Third-generation CARs were defined by the inclusion of a second costimulatory signaling domain intended to improve even further the antitumor ability of the cells, most commonly combining CD28 with 4-1BB (1517). However, the clinical advantages of using third- compared with second-generation CAR T cells are unclear.

In a recent clinical trial, second- and third-generation CD19-specific CAR T cells were coadministered to patients with B cell non-Hodgkin’s lymphoma, demonstrating that third-generation cells had improved in vivo expansion and persistence (18). The study and selection of an optimal costimulatory domain continues to be an expanding field in CAR T cell biology and includes OX40 (19), ICOS (20), CD27 (21), and others (22). The evolution into different generations of CAR T cells is based on harnessing immunological traits of T cells that are beneficial for an enhanced antitumor response. These include optimal cell activation received through signal 1 (TCR/CD3ζ) and signal 2 (costimulatory receptors) that will affect their effector function. For cell therapy, persistence after adoptive transfer is essential for durable responses, with a less differentiated phenotype being a key trait for enhanced engraftment and subsequent expansion after antigenic stimulation (23, 24). Optimizing designs of these different components of the CAR and/or T cells has the potential for improving clinical outcomes by addressing challenges identified from the early clinical application of CAR T cells.

Toxicities associated with CAR T cell administration

Cytokine release syndrome (CRS) and neurotoxicity are commonly observed toxicities after the administration of CAR T cells (25). While exact pathophysiology of these syndromes is not yet fully elucidated, one proposed mechanism is that systemic cytokines can be released when there is an interaction between the tumor and the CAR T cells (26). CRS manifests with fevers, hypoxia, and hypotension, which can lead to multiorgan dysfunction (25). Neurologic toxicity, termed immune effector cell–associate neurotoxicity syndrome (ICANS), can present with aphasia, altered level of consciousness, impaired cognitive function, motor weakness, seizures, and/or cerebral edema (25, 27). It is important to note that comparing toxicities across CAR T trials is challenging, as there is variability in the scoring systems used to grade the severity of toxicities (Table 1).

Table 1. Summary of select clinical trials highlighting various mechanisms to reduce CAR T cell toxicity and improve efficacy.

graphic file with name jci-131-142030-g114.jpg

Although toxicities are usually reversible with supportive care and/or interventions such as corticosteroids or IL-6 receptor blockade, they can be fatal, highlighting the need for safer CAR T constructs that maintain antitumor ability (28). Current guidelines support the use of anti–IL-6 therapy in combination with corticosteroids for patients with grade 3 or higher CRS and/or neurotoxicity. In an analysis of the ZUMA-1 trial, prophylactic use of tocilizumab reduced the incidence of severe CRS from 13% to 5%, but did not reduce the incidence of neurotoxicity (29). Topp and colleagues reported that earlier intervention with tocilizumab and steroids in patients enrolled in cohort 4 of the ZUMA-1 trial improved the incidence of severe CRS and neurotoxicity without negatively affecting clinical outcomes (30). This study and others challenge the notion that early steroid intervention hinders CAR T cell expansion (28). Prospective studies are needed to address whether prophylactic cytokine blockade and/or corticosteroids reduce the incidence of severe toxicity without affecting CAR T cell expansion and clinical efficacy.

In order for infused CAR T cells to be therapeutically efficient, they need to reach an activation threshold that is influenced by antigen density on the target tissue, affinity of the antigen-binding site to said antigen (31), costimulatory domains present in the CAR, and other cell-intrinsic and -extrinsic factors (32). It was recently shown that low expression levels of CD22 on target cells modified CD22-specific CAR T cell function, with lower cytokine production and reduced persistence in mouse models of acute lymphoblastic leukemia (ALL) with low antigen expression (33). Another study demonstrated that the activity of CD19-specific CAR T cells depends on the antigen density on target cells, with constructs including CD28 costimulation showing better function against low-antigen tumor cells than 4-1BB constructs. This lower sensitivity is enhanced by including additional ITAM motifs to the 4-1BB–based CAR and shows increased signaling against low antigen density (34). The costimulatory domain was shown to modulate the activity of CAR T cells, with CD28 inducing more rapid responses that were higher in magnitude compared with 4-1BB–containing CARs, with lower risks of cytokine-associated toxicities (Figure 2A and ref. 35). The choice of costimulatory domain affects not only CAR T cell fate; in addition, the length of the endodomain can also modulate activation. It was demonstrated that a shorter intracellular domain containing CD28 as costimulation reduced the ability of the CAR to interact with CD3ζ, with consequently less cell activation and cytokine production (Figure 2B and ref. 36). In another study, an anti-CD19 CAR with a longer hinge/transmembrane and intracellular fragments produced lower levels of cytokines with higher expression levels of antiapoptotic proteins with cytolytic ability comparable to that of the conventional 4-1BB–based CAR with shorter domains. A phase I clinical trial using this construct showed no neurotoxicity or CRS in any of the 25 treated patients (Figure 2C and ref. 37).

Figure 2. Overcoming toxicities through CAR-engineering strategies.

Figure 2

CAR T cells show different levels of activation and cytokine production depending on (A) costimulatory domain, (B and C) length of the transmembrane domain, and (D) nature and composition of the hinge/transmembrane domain (E). CAR T cells have been engineered to secrete soluble IL-1 receptor antagonists or induce a targeted deletion on the gene encoding for GM-CSF associated with toxicities (F). Administration of a protease inhibitor controls CAR surface expression, acting as an “on/off switch” based on a complex composed of a self-cleaving protease and a degron moiety. TM, transmembrane; AICD, activation-induced cell death.

The transmembrane domain also modifies the ability of the CAR to induce cell activation. It was shown that an anti-CD19 CAR with a transmembrane moiety containing CD8α produced fewer cytokines and lower levels of activation-induced cell death compared with a CD28-based transmembrane with comparable ability to eliminate tumors in preclinical models (Figure 2D and ref. 38). Based on these findings, a phase I clinical trial utilizing a fully humanized anti-CD19 antibody plus hinge and transmembrane domains from CD8α (Hu19-CD828Z) was designed to test the hypothesis that single-chain variable fragments (scFvs) derived from a human antibody may be less immunogenic than those from a murine-derived antibody. Twenty patients with large B cell lymphoma (LBCL) were enrolled in this study with ongoing complete remission (CR) rates of 40%, comparable to that in the pivotal ZUMA-1 study. The incidence of severe grade 3 or higher neurologic toxicity was only 5% compared with 32% in the ZUMA-1 trial, with lower serum levels of cytokines compared with those of patients treated with an earlier construct, FMC63-28Z (4). This work demonstrated that newer generation CARs associated with lower levels of in vitro cytokine production are effective yet safer than prior CAR constructs (39).

One approach to mitigating CRS is to genetically delete cytokines associated with toxicities or to induce the secretion of molecules that inhibit their biological action, such as soluble cytokine receptors. The cytokine GM-CSF has recently been associated with the development of CRS. In a lymphoblastic leukemia xenograft model, the neutralization of GM-CSF by lenzilumab reduced neurotoxicity. The deletion of GM-CSF specifically on CAR T cells improved the antitumor activity and survival of mice (40). Based on these results, a clinical trial evaluating the combination of lenzilumab with axicabtagene ciloleucel (axi-cel) in patients with relapsed/refractory (R/R) LBCL is planned (NCT04314843). Similarly, IL-1 secreted by macrophages/monocytes has been shown to play a key role in CRS, and this effect can be countered by engineering CAR T cells able to secrete IL-1 receptor antagonist (IL-1Ra) (Figure 2E and ref. 41) or by administering anakinra, a soluble form of IL-1Ra (42). A phase II clinical trial is accruing patients to evaluate the efficacy of anakinra in reducing severe CRS and neurotoxicity in patients with LBCL treated with axi-cel (NCT04150913).

On/off switches by administration of exogenous small molecules.

A novel strategy for controlling adverse events arising from CAR T cell therapy focuses on depleting or arresting infused cells by including safety mechanisms such as suicide genes or safety switches. One approach engineers cells able to engage full antitumor potential upon administration of bispecific adaptors that act as bridges between tumor cells and CAR T cells. These low molecular weight adaptors contain residues able to recognize molecules expressed by cancer cells, such as folate receptors, to engage the CAR-inducing cell activation. Consequently, CRS can then be controlled and even terminated by regulating the administration of the bispecific adaptor (43).

Another strategy for reducing severe toxicities is to selectively trigger apoptosis by introducing an inducible form of caspase 9 (iCasp9) that dimerizes upon exposure to an inert small molecule (AP1903). This approach was initially reported in pediatric patients with acute leukemia who underwent stem cell transplantation, and it led to a rapid deletion of infused T cells (44). This was further evaluated in a humanized preclinical model of CD19-specific CAR T cells with depletion of transferred cells and subsequent B cell reconstitution. The effect was dependent on the dose of dimerizing agent administered (45). A caveat of using suicide genes is the corresponding decrease in antitumor activity, and therefore these strategies may be best utilized in cases of life-threatening toxicity (46). Robust clinical data regarding suicide genes is lacking. Several phase I clinical trials are planned to determine the safety and tolerability of iCasp9 CAR T cells in patients with CD19-expressing B cell malignancies (NCT03016377, NCT03696784).

Investigators evaluated selective expression of the CAR in the cell surface by engineering CAR constructs with an “off switch” encoded as part of the receptor. In this design, a self-cleaving site is controlled by a protease paired to a “degron” moiety (a degradation signal) that is able to induce the proteolysis of the CAR protein. In the “on” state, the self-cleavage will generate a CAR able to be expressed on the cell surface, while the degron will undergo proteolysis. The administration of a protease inhibitor (asunaprevir) switches to the “off” state in which the CAR is not cleaved and retains the degron moiety that induces proteasomal degradation of the CAR-degron protein, thus preventing cell-surface expression (Figure 2F and ref. 47).

Dasatinib, a tyrosine kinase inhibitor, disrupts signaling downstream of the activation domain CD3ζ by inhibiting phosphorylation of the lymphocyte-specific protein tyrosine kinase (LCK). Therefore, dasatinib temporarily inactivates CAR T cells in vitro and in vivo, with full antitumor effect restored upon removal of the drug. In a preclinical model of lymphoma, the administration of dasatinib rapidly mitigated CRS (48). Hence, as dasatinib targets the activation motif CD3ζ, it could be used to rapidly reduce CRS of CAR T cells independently of their costimulatory domains.

Small molecular inhibitors can be used to increase the efficacy of CAR T cells. Bruton’s tyrosine kinase (BTK) is highly expressed in B cell malignancies, and its inhibitor ibrutinib has been used to treat these malignancies. In a combinatorial approach with CAR T cell therapy, it was shown that ibrutinib does not alter the gene transfer of CD19-CAR T cells, their proliferation, or their in vitro cytotoxic ability. Combining anti-CD19 CAR T and ibrutinib increased engraftment and antitumor efficacy on in vivo models of ALL and chronic lymphocytic leukemia (CLL) (49). It was recently reported that the concomitant administration of CD19-CAR T cells and ibrutinib in patients with CLL showed lower severity of CRS with expansion similar to that of CAR T cells alone (50).

Allogeneic CARs.

Despite the clinical efficacy observed with autologous CAR T cell therapy for hematologic malignancies, the complex manufacturing period and financial burden of treatment may limit the widespread use of these therapies. It was recently shown that patients who required bridging therapy between the time of apheresis and CAR T cell infusion had worse overall response. Furthermore, in the US CAR T Cell Lymphoma Consortium of patients with R/R LBCL, 7% of patients did not receive axi-cel after apheresis due to progression and/or death, highlighting the downside of waiting for the manufacturing of cell therapy products (51). One way to mitigate this challenge is by developing “off the shelf” allogeneic CAR T products. Nonetheless, the use of allogeneic T cells can give rise to graft-versus-host disease (GVHD). In order to mitigate this, cells have been modified to disrupt the TCR gene and/or the HLA I gene (52). Some groups have employed CRISPR-Cas9 to knock out genes such as TRAC, B2M, and HLA class I for CD19-specific allogeneic CAR T cells maintaining antitumor activity (53, 54). However, the safety and efficacy of this approach may be affected by a proportion of cells still expressing the intended knocked out genes.

Compared with T cells, allogeneic NK cells have the benefit of being relatively safe to infuse even without complete HLA matching, as they do not express a TCR. However, their low persistence after administration limits their therapeutic efficacy (55). In a phase I trial of allogeneic CD19-directed CAR NK cells derived from cord blood, 8 out of 11 patients showed response (56). Notably, none of the patients developed CRS, neurotoxicity, or GVHD, and the maximum tolerated dose was not reached (56). Although the numbers in this study are small, the capability of producing a safe off-the-shelf product can increase access to treatment. Ongoing clinical trials will inform the durability of response and toxicity profile of allogeneic CAR T cell therapies.

Antigen escape

Despite the high response rates of CD19 CAR T cell therapy, the loss or downregulation of CD19 has been reported as a common mechanism of tumor resistance (57, 58). Antigenic escape has been attributed to mutations and splice variants of CD19, leading to low or absent expression of the protein on the surface of malignant cells (59). A recently described mechanism mediating antigen escape consists of the transfer of the target antigen from the tumor cell to the CAR T by a mechanism of trogocytosis. This process of antigen transfer is dependent on CAR engagement to its target, leading to a reduction in the antigen density on the tumor cell and subsequent fratricide. This phenomenon was observed in CAR T cells specific for CD19, CD22, B cell maturation antigen (BCMA), and mesothelin (60).

Current strategies focused on overcoming antigen escape include the use of CAR T cells able to recognize more than one antigen present in the tumor cell. The use of multi-targeted CAR T cell products is an attractive approach that can be obtained by mixing single-targeted cells against different antigens prior to infusion (coadministration) or by simultaneously transducing T cells with different CAR constructs (cotransduction). Another multi-targeted strategy involves employing a single CAR molecule able to recognize two antigens. A bicistronic construct involves one single vector encoding for two independent CAR molecules, while bispecific constructs encode for a single bivalent CAR molecule able to recognize two different antigens (61).

The administration of bispecific CAR T cells able to recognize CD19 and CD20 demonstrated enhanced antitumor activity in preclinical models of B cell malignancies compared with a single CD19 construct. This effect was attributed to a reduced antigen escape by tumor cells (62). In another study, bispecific CAR T cells engineered against both CD19 and CD22 showed in vitro antitumor activity comparable to that of monospecific CD19 CAR T cells, but were able to eradicate CD19-negative patient-derived xenografts (63).

While several groups are evaluating dual-targeted CAR T cells in clinical trials as a strategy for overcoming antigen escape, data are not yet mature enough to determine whether these approaches affect clinical outcomes. Another unique strategy is the sequential infusion of two third-generation CARs targeting CD19 and CD22, respectively. In a single-center study of 89 patients with R/R B cell malignancies, Pan et al. demonstrated the feasibility of this approach and potential for clinical benefit (64). However, patients who relapsed did have evidence of antigen escape with loss of CD19. The use of two different cell populations raises concerns of additive toxicity. While this approach could work in hematologic malignancies, it would be challenging to translate to solid tumors in which there is much more heterogenous antigen expression.

A strategy focused on targeting multiple molecules includes engineering CAR T cells to produce bi-specific T cell engagers (BiTEs). BiTEs are soluble molecules that can be secreted and consist of two scFvs: one able to ligate CD3 and the other to bind a desired molecule expressed on the tumor cell. Therefore, BiTEs can act as physical links between T cells and tumor cells. A CD19-specific BiTE, blinatumomab, was granted approval in 2018 by the FDA for the treatment of B cell precursor ALL (B-ALL) (65). Engineering CAR T cells able to engage and enhance the endogenous immune system of the patient by secreting BiTEs is one approach that can enhance an antitumor T cell response, especially in patients with solid tumors where there is heterogenous antigen expression. A recent report found that CAR T cells able to secrete BiTEs can overcome antigen escape in preclinical models of B-ALL and circumvent antigen heterogeneity in a solid model of glioblastoma (66).

Solid tumors

Despite the impressive clinical results observed in hematological malignancies, progress for CAR T cell therapy in solid tumors has been met with many obstacles. One major factor is the lack of tumor-specific cell-surface antigens in solid tumors. The biologic heterogeneity in solid tumors differs from that in B cell hematologic malignancies and leads to high incidence of “on-target/off-tumor” toxicities due to the expression of target antigens on normal healthy tissues. Moreover, the complex TME in solid tumors negatively affects both endogenous T cells and transferred CAR T cells. This immune-suppressive microenvironment composed of soluble factors and suppressive cells such as myeloid-derived suppressor cells (MDSCs) and Tregs, in addition to the physical barriers that inhibit T cell infiltration into the tumor, add to the challenges for successful CAR T development for solid tumors.

Identifying a tumor-associated antigen in solid tumors.

In contrast to B cell malignancies, solid tumors have heterogeneous tumor antigen expression, and these antigens can be expressed in low levels on normal tissues. Even low-level expression in healthy tissues can lead to fatal events (67, 68). Preclinical approaches to improving safety in targeting antigens found in healthy tissue include development of CARs with lower affinity for target proteins, which demonstrated less exhaustion and enhanced proliferation in vivo (69). Several groups have used immunoproteomics, DNA or RNA sequencing, and whole gene exome sequencing to identify novel tumor-associated antigens and neoantigens, including prostate-specific membrane antigen (PSMA) and prostate stem cell antigen (PSCA). Kloss et al. reported a strategy for enhancing tumor specificity by combinatorial recognition of two antigens, PSMA and PSCA, in a preclinical model. The novelty of this approach was that one CAR had a CD28-signaling domain while the other had a CD3ζ-signaling domain. This led to an improved toxicity profile by reducing reactivity against healthy tissues expressing either antigen alone (70).

Another strategy includes synthetic Notch receptors that were developed to conditionally express the CAR upon cognate ligand engagement on the tumor site. This dual antigenic trigger controls CAR expression in a transcriptional level that can mitigate systemic toxicities and expand the antigen repertoire (71, 72). Efforts are being made to broaden the antigen-binding domain of CARs beyond scFvs. Some approaches include molecules with a lower molecular size, such as camelid single-domain antibody fragments or “nanobodies” (73), ankyrin repeats (DARPins) (74, 75), and the recently described D-domain CARs. In a recent study, T cells armed with a nanobody targeting CD13 were shown to induce antitumor response against preclinical models of myeloid leukemia (76). Moreover, D-domains also have a smaller size than scFvs. These molecules are single-domain structures derived from α-helical bundle protein αD3 able to recognize antigens. In another recent study, T cells armed with a CD123-specific D-domain showed durable antitumor activity in xenograft models of acute myeloid leukemia (77).

Delivery and trafficking to the tumor site.

In order for CAR T cells to target surface antigens in solid tumors, they must first traffic to the tumor site and subsequently infiltrate the tumor. To overcome this challenge, local administration of CAR T cells into the tumor site is an approach used to increase the number of available cells tested in brain (78), breast (79), peritoneal carcinomatosis (80), head and neck (81), liver metastasis (82), and lung (83). Although this approach showed promising results, it may be limited to certain types of tumors and could have limited efficacy against distal lesions from the inoculated primary site.

Different chemokines mediate trafficking of immune cells to the tumor bed (84). To exploit this feature, CAR T cells have been engineered to express chemokine receptors in order to increase trafficking to the tumor site (Figure 3A). The coexpression of CC-chemokine receptor 2 (CCR2), receptor for CCL2, together with a CAR specific for the target antigen GD2 was shown to improve trafficking and tumor infiltration in a xenograft model of neuroblastoma (85). Another study demonstrated that expressing CCR2 together with a mesothelin-specific CAR enhanced tumor infiltration compared with CAR T cells with no expression of the chemokine receptor (86). Using another chemokine receptor, CXC-chemokine receptor 2–expressing (CXCR2-expressing) CAR T cells directed against αvβ6 were able to control tumor growth with higher efficacy than the conventional counterpart in xenograft models of solid tumors (87).

Figure 3. Strategies to enhance efficacy of CAR T cells against solid tumors.

Figure 3

Several engineering strategies have been developed to improve CAR T cell function (A). Chemokine receptors coexpressed on CAR T cells enhance traffic to the tumor site (B). Expression of heparanase improves the ability of CAR T cells to penetrate the tumor stoma by degrading the ECM (C). Engineered CAR T cells can overcome inhibitory immune checkpoints by secreting anti–PD-1 antibodies or scFvs; another strategy includes the expression of dominant-negative forms of the receptors (DN–PD-1) (D). CAR T cells modified to secrete cytokines have improved function by modulating the tumor milieu. CCR4, CC-chemokine receptor 4; DNR, dominant-negative receptor.

In order to enhance the penetration ability of CAR T cells into the tumor, cells can be engineered to produce and secrete enzymes that degrade the extracellular matrix (ECM) and thus facilitate tumor infiltration. GD2-specific CAR T cells transduced to overexpress the enzyme heparanase improved the ability of transferred cells to degrade ECM, promoting tumor infiltration and antitumor activity in xenograft models (Figure 3B and ref. 88). In one study, the coexpression of IL-7 and CCL19 in CAR T cells improved the tumor-infiltrating ability of these cells, showing regression of preestablished solid tumors in mouse models (89). Another study reported that nanobody-based CAR T cells directed against molecules highly expressed in the TME, such as PD-L1 and the EIIIB splice variant of fibronectin, showed tumor growth inhibition in solid tumors engrafted in immunocompetent mice (90).

Overcoming the TME.

The microenvironment within solid tumors is immunosuppressive due in part to the presence of cells with immune-suppressive activity and tumor-derived cytokines as well as checkpoint inhibitory ligands (91). CAR T cells can be engineered to be resistant to immune checkpoints or secrete scFvs mimicking systemic administration of these therapeutic antibodies (Figure 3C). In one study, CAR T cells modified to secrete anti–programmed cell death 1 (anti–PD-1) scFvs have been shown to act in a paracrine and autocrine manner, enhancing the antitumor activity of both the transferred CAR T cells and the endogenous T cells (92). In a similar approach, CAR T cells engineered to secrete an anti–PD-L1 antibody showed improved antitumor function in a preclinical model of renal cell carcinoma (93). These results were achieved in xenografts and immunocompetent mouse models by blocking the PD-1/PD-L1 axis in situ.

Another approach focused on this axis includes CAR T cells cotransduced to express a dominant-negative form of PD-1 lacking the intracellular signaling domain of the receptor. The inclusion of this truncated form of PD-1, unable to transduce inhibitory signals, improved the antitumor activity of mesothelin-specific CAR T cells in a xenograft model of mesothelioma (94). Other studies have reported that by disrupting the endogenous expression of PD-1 on CAR T cells by CRISPR/Cas9, the antitumoral activity of the transferred cells is enhanced in different mouse models (9597). It was recently demonstrated that exosomes derived from CAR T cells express CAR molecules and carry cytotoxic molecules that showed antitumor activity upon injection into models of mice bearing solid tumors. These CAR-containing exosomes do not express PD-1, as opposed to CAR T cells, and their in vivo antitumor function is not affected by PD-L1 engagement in the TME (98). Clinical trials with PD-1 knockout autologous CAR T cells are underway for esophageal cancer (NCT03706326), lung cancer (NCT03525782), ALL (NCT03298828), and mesothelin-expressing tumors (NCT03545815).

Few early phase clinical trials have evaluated the efficacy of combining CAR T cells with checkpoint inhibitors. The phase 1/2 ZUMA-6 trial investigated the combination of axi-cel with the anti–PD-L1 antibody atezolizumab in patients with R/R LBCL (NCT0926833). Although there were a small number of patients (n = 28), the best objective response rate of 75% (46% CR) is encouraging. The incidence of CRS and neurotoxicity was comparable to that in the ZUMA-1 trial (99). In patients with R/R neuroblastoma, the addition of pembrolizumab to CAR T cell therapy did not enhance persistence. However, no conclusions can be drawn, as only three patients were treated in this cohort (100). In another report, patients with malignant pleural disease were treated with regionally delivered mesothelin-targeted CAR T cells. Fourteen of the patients received checkpoint blockade agents off protocol and did not experience any toxicity (101). Therefore, the second phase of this trial is evaluating the efficacy of adding pembrolizumab after CAR T infusion (NCT02414269).

Modifying the intratumoral cytokine pool.

In order to modulate the suppressive soluble milieu present in solid tumors, CAR T cells can be engineered to secrete immunostimulatory cytokines that enhance activation, proliferation, and antitumor activity (Figure 3D). Some studies showed that CAR T cells designed to secrete IL-12 have enhanced antitumor function in preclinical models of solid tumors, with an additional stimulation of an endogenous antitumor T cell response (102105). This same beneficial effect of IL-12–secreting CAR T cells was observed for B cell malignancies (106). Another cytokine involved in T cell homeostasis and survival is IL-15. It was demonstrated that CAR T cells armored to secrete IL-15 were able to eradicate more efficiently a xenogeneic metastatic model of neuroblastoma (107). In a similar report, IL-13Rα2–specific CAR T cells engineered to produce IL-15 showed increased in vivo persistence and antitumor activity against glioblastoma models compared with the conventional counterpart (108). Furthermore, CAR T cells designed to inducibly secrete IL-18 showed superior antitumor activity against preclinical pancreatic and lung tumors. Importantly, an increase in numbers of intratumoral M1 macrophages and NK cells, which are associated with antitumor response, was reported for IL-18–armored CAR T cells, while protumoral populations such as Tregs and M2-like macrophages were reduced (109). In a similar study, both human and murine IL-18–secreting CAR T cells showed enhanced proliferative capacity and antitumor immunity in mouse models of solid tumors (110).

Resisting immunosuppressive cells.

The presence of immunosuppressive cells in the TME, such as Tregs, MDSCs, and M2-like macrophages, has been shown to inhibit the antitumor ability of tumor-infiltrating T cells (111, 112). It has been demonstrated that these cells also inhibit the antitumor function of transferred CAR T cells in solid tumor models (113). An approach to limiting this suppression is the administration of CAR T cells in combination with agents that reduce the number of these immunosuppressive cells. A study showed that pediatric sarcoma xenografts induced the accumulation of murine MDSCs that inhibited GD2-specific CAR T cell function. The coadministration of GD2-CAR T cells with all-trans retinoic acid reduced the number of MDSCs and improved the antitumor ability of the transferred cells in this model of sarcoma (114). In another report, the transfer of Her2-specific CAR T cells in combination with an anti–4-1BB antibody enhanced the antitumor function of CAR T cells and showed reduced numbers of Tregs and MDSCs in a preclinical orthotopic model of breast cancer (115). Harnessing the capacity of macrophages to penetrate tumors, investigators demonstrated that genetically engineered human macrophages with CARs (CAR-Ms) decreased tumor burden and prolonged survival in xenograft mouse models. Furthermore, CAR-Ms remodeled the TME and were capable of antigen presentation in humanized mouse models (116).

Although chemotherapy-based lymphodepletion reduces the number of immunosuppressive cells to favor the engraftment of transferred cells (100, 117), the idea of engineering CAR T cells that are able to resist the action of these immunosuppressive cells is promising. It was recently demonstrated that introducing mutations at the LCK-binding motif in the CD28 intracellular domain of EGFRvIII-targeting CAR T cells improved antitumor activity against a melanoma tumor model. These mutations rendered CAR T cells resistant to suppression by Tregs without the need for lymphodepleting schemes (118). In another study, VEGFR-2–targeted CAR T cells reduced the number of a subset of intratumoral MDSCs that expressed VEGFR-2 in different models of vascularized subcutaneous tumors (119). These approaches focused on reducing the number of immunosuppressive cells in the TME or rendering CAR T cells resistant to this suppression, which could improve the therapeutic efficacy of this treatment for solid malignancies.

Concluding remarks

CAR T cell therapy has shown remarkable clinical results against B cell malignancies, with early indications that its use for solid tumors could pose a new therapeutic option. Promising bioengineering strategies are being developed to reduce CAR T cell–associated toxicities and improve therapeutic efficacy. Here, we reviewed preclinical and clinical approaches employed to mitigate toxicities and overcome common mechanisms of tumor evasion. Furthermore, we discussed major barriers imposed by solid tumors together with strategies that can enhance and broaden the therapeutic application of CAR T cells in these scenarios. Data obtained from both preclinical research on CAR design optimization and clinical trials are key to shaping next-generation CAR therapeutics with superior outcomes for cancer patients.

Version 1. 01/19/2021

Print issue publication

Footnotes

Conflict of interest: MLD and RF received research funding from Novartis. MLD receives research funding from Precigen, Atara Biotherapeutics, and ELICIO Therapeutics. MLD has received licensing fees from Atara Biotherapeutics for CAR intellectual property. MLD has equity in Precision Biosciences and Adaptive Biotechnologies and unexercised options with Bellicum and CERtainty.

Copyright: © 2021, American Society for Clinical Investigation.

Reference information: J Clin Invest. 2021;131(2):e142030.https://doi.org/10.1172/JCI142030.

Contributor Information

Emiliano Roselli, Email: Emiliano.Roselli@moffitt.org.

Rawan Faramand, Email: Rawan.Faramand@moffitt.org.

Marco L. Davila, Email: marco.davila@moffitt.org.

References

  • 1.Sanmamed MF, Chen L. A paradigm shift in cancer immunotherapy: from enhancement to normalization. Cell. 2018;175(2):313–26. doi: 10.1016/j.cell.2018.09.035. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 2.Rosenberg SA, Restifo NP. Adoptive cell transfer as personalized immunotherapy for human cancer. Science. 2015;348(6230):62–68. doi: 10.1126/science.aaa4967. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 3.Roselli E, et al. CAR-T engineering: optimizing signal transduction and effector mechanisms. BioDrugs. 2019;33(6):647–659. doi: 10.1007/s40259-019-00384-z. [DOI] [PubMed] [Google Scholar]
  • 4.Neelapu SS, et al. Axicabtagene ciloleucel CAR T-cell therapy in refractory large B-cell lymphoma. N Engl J Med. 2017;377(26):2531–2544. doi: 10.1056/NEJMoa1707447. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 5.Ruella M, Kenderian SS. Next-generation chimeric antigen receptor T-cell therapy: going off the shelf. BioDrugs. 2017;31(6):473–481. doi: 10.1007/s40259-017-0247-0. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 6.Locke FL, et al. Long-term safety and activity of axicabtagene ciloleucel in refractory large B-cell lymphoma (ZUMA-1): a single-arm, multicentre, phase 1–2 trial. Lancet Oncol. 2019;20(1):31–42. doi: 10.1016/S1470-2045(18)30864-7. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 7.Schuster SJ, et al. Tisagenlecleucel in adult relapsed or refractory diffuse large B-cell lymphoma. N Engl J Med. 2019;380(1):45–56. doi: 10.1056/NEJMoa1804980. [DOI] [PubMed] [Google Scholar]
  • 8.Rafiq S, et al. Engineering strategies to overcome the current roadblocks in CAR T cell therapy. Nat Rev Clin Oncol. 2020;17(3):147–167. doi: 10.1038/s41571-019-0297-y. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 9.Gross G, et al. Expression of immunoglobulin-T-cell receptor chimeric molecules as functional receptors with antibody-type specificity. Proc Natl Acad Sci U S A. 1989;86(24):10024–10028. doi: 10.1073/pnas.86.24.10024. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 10.Eshhar Z, et al. 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 U S A. 1993;90(2):720–724. doi: 10.1073/pnas.90.2.720. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 11.Kershaw MH, et al. A phase I study on adoptive immunotherapy using gene-modified T cells for ovarian cancer. Clin Cancer Res. 2006;12(20):6106–6115. doi: 10.1158/1078-0432.CCR-06-1183. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 12.Krause A, et al. Antigen-dependent CD28 signaling selectively enhances survival and proliferation in genetically modified activated human primary T lymphocytes. J Exp Med. 1998;188(4):619–626. doi: 10.1084/jem.188.4.619. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 13.Imai C, et al. Chimeric receptors with 4-1BB signaling capacity provoke potent cytotoxicity against acute lymphoblastic leukemia. Leukemia. 2004;18(4):676–684. doi: 10.1038/sj.leu.2403302. [DOI] [PubMed] [Google Scholar]
  • 14.van der Stegen SJC, et al. The pharmacology of second-generation chimeric antigen receptors. Nature Reviews Drug Discovery. 2015;14(7):499–509. doi: 10.1038/nrd4597. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 15.Carpenito C, et al. Control of large, established tumor xenografts with genetically retargeted human T cells containing CD28 and CD137 domains. Proc Natl Acad Sci U S A. 2009;106(9):3360–3365. doi: 10.1073/pnas.0813101106. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 16.Milone MC, et al. Chimeric receptors containing CD137 signal transduction domains mediate enhanced survival of T cells and increased antileukemic efficacy in vivo. Mol Ther. 2009;17(8):1453–1464. doi: 10.1038/mt.2009.83. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 17.Zhong X-S, et al. Chimeric antigen receptors combining 4-1BB and CD28 signaling domains augment PI3kinase/AKT/Bcl-XL activation and CD8+ T cell-mediated tumor eradication. Mol Ther. 2010;18(2):413–420. doi: 10.1038/mt.2009.210. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 18.Ramos CA, et al. In vivo fate and activity of second- versus third-generation CD19-specific CAR-T cells in B cell non-hodgkin’s lymphomas. Mol Ther. 2018;26(12):2727–2737. doi: 10.1016/j.ymthe.2018.09.009. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 19.Hombach AA, et al. OX40 costimulation by a chimeric antigen receptor abrogates CD28 and IL-2 induced IL-10 secretion by redirected CD4(+) T cells. Oncoimmunology. 2012;1(4):458–466. doi: 10.4161/onci.19855. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 20.Shen C-J, et al. Chimeric antigen receptor containing ICOS signaling domain mediates specific and efficient antitumor effect of T cells against EGFRvIII expressing glioma. J Hematol Oncol. 2013;6:33. doi: 10.1186/1756-8722-6-33. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 21.Song D-G, et al. CD27 costimulation augments the survival and antitumor activity of redirected human T cells in vivo. Blood. 2012;119(3):696–706. doi: 10.1182/blood-2011-03-344275. [DOI] [PubMed] [Google Scholar]
  • 22.Weinkove R, et al. Selecting costimulatory domains for chimeric antigen receptors: functional and clinical considerations. Clin Transl Immunology. 2019;8(5):e1049. doi: 10.1002/cti2.1049. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 23.Fraietta JA, et al. Determinants of response and resistance to CD19 chimeric antigen receptor (CAR) T cell therapy of chronic lymphocytic leukemia. Nat Med. 2018;24(5):563–571. doi: 10.1038/s41591-018-0010-1. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 24.Feucht J, et al. Calibration of CAR activation potential directs alternative T cell fates and therapeutic potency. Nature Medicine. 2019;25(1):82–88. doi: 10.1038/s41591-018-0290-5. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 25.Lee DW, et al. ASBMT consensus grading for cytokine release syndrome and neurologic toxicity associated with immune effector cells. Biol Blood Marrow Transplant. 2019;25(4):625–638. doi: 10.1016/j.bbmt.2018.12.758. [DOI] [PubMed] [Google Scholar]
  • 26.Faramand RG, et al. Tumor microenvironment composition and severe cytokine release syndrome (CRS) influence toxicity in patients with large B cell lymphoma treated with axicabtagene ciloleucel. Clin Cancer Res. 2020;26(18):4823–4831. doi: 10.1158/1078-0432.CCR-20-1434. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 27.Neelapu SS, et al. Chimeric antigen receptor T-cell therapy - assessment and management of toxicities. Nat Rev Clin Oncol. 2018;15(1):47–62. doi: 10.1038/nrclinonc.2017.148. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 28.Davila ML, et al. Efficacy and toxicity management of 19-28z CAR T cell therapy in B cell acute lymphoblastic leukemia. Sci Transl Med. 2014;6(224):224ra225. doi: 10.1126/scitranslmed.3008226. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 29.Locke FL, et al. Preliminary results of prophylactic tocilizumab after axicabtageneciloleucel (axi-cel; KTE-C19) treatment for patients with refractory, aggressive non-hodgkin lymphoma (NHL) Blood. 2017;130(Suppl 1):1547 [Google Scholar]
  • 30. Topp M, et al. Earlier steroid use with axicabtagene ciloleucel (axi-cel) in patients with relapsed/refractory large B cell lymphoma. Presented at: 60th American Society of Hematology Annual Meeting; December 1–4, 2018; San Diego, CA 2019. https://doi.org/10.1182/blood-2019-126081.
  • 31.Park S, et al. Micromolar affinity CAR T cells to ICAM-1 achieves rapid tumor elimination while avoiding systemic toxicity. Sci Rep. 2017;7(1):14366. doi: 10.1038/s41598-017-14749-3. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 32.Milone MC, Bhoj VG. The pharmacology of T cell therapies. Mol Ther Methods Clin Dev. 2018;8:210–221. doi: 10.1016/j.omtm.2018.01.010. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 33.Ramakrishna S, et al. Modulation of target antigen density improves CAR T-cell functionality and persistence. Clin Cancer Res. 2019;25(17):5329–5341. doi: 10.1158/1078-0432.CCR-18-3784. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 34.Majzner RG, et al. Tuning the antigen density requirement for CAR T-cell activity. Cancer Discov. 2020;10(5):702–723. doi: 10.1158/2159-8290.CD-19-0945. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 35.Salter AI, et al. Phosphoproteomic analysis of chimeric antigen receptor signaling reveals kinetic and quantitative differences that affect cell function. Sci Signal. 2018;11(544):eaat6753. doi: 10.1126/scisignal.aat6753. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 36.Ramello MC, et al. An immunoproteomic approach to characterize the CAR interactome and signalosome. Sci Signal. 2019;12(568):eaap9777. doi: 10.1126/scisignal.aap9777. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 37.Ying Z, et al. A safe and potent anti-CD19 CAR T cell therapy. Nat Med. 2019;25(6):947–953. doi: 10.1038/s41591-019-0421-7. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 38.Alabanza L, et al. Function of novel anti-CD19 chimeric antigen receptors with human variable regions is affected by hinge and transmembrane domains. Mol Ther. 2017;25(11):2452–2465. doi: 10.1016/j.ymthe.2017.07.013. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 39.Brudno JN, et al. Safety and feasibility of anti-CD19 CAR T cells with fully human binding domains in patients with B-cell lymphoma. Nat Med. 2020;26(2):270–80. doi: 10.1038/s41591-019-0737-3. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 40.Sterner RM, et al. GM-CSF inhibition reduces cytokine release syndrome and neuroinflammation but enhances CAR-T cell function in xenografts. Blood. 2019;133(7):697–709. doi: 10.1182/blood-2018-10-881722. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 41.Giavridis T, et al. CAR T cell-induced cytokine release syndrome is mediated by macrophages and abated by IL-1 blockade. Nat Med. 2018;24(6):731–738. doi: 10.1038/s41591-018-0041-7. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 42.Norelli M, et al. Monocyte-derived IL-1 and IL-6 are differentially required for cytokine-release syndrome and neurotoxicity due to CAR T cells. Nat Med. 2018;24(6):739–748. doi: 10.1038/s41591-018-0036-4. [DOI] [PubMed] [Google Scholar]
  • 43.Lee YG, et al. Regulation of CAR T cell-mediated cytokine release syndrome-like toxicity using low molecular weight adapters. Nat Commun. 2019;10(1):2681. doi: 10.1038/s41467-019-10565-7. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 44.Di Stasi A, et al. Inducible apoptosis as a safety switch for adoptive cell therapy. N Engl J Med. 2011;365(18):1673–1683. doi: 10.1056/NEJMoa1106152. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 45.Diaconu I, et al. Inducible caspase-9 selectively modulates the toxicities of CD19-specific chimeric antigen receptor-modified T cells. Mol Ther. 2017;25(3):580–592. doi: 10.1016/j.ymthe.2017.01.011. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 46.Paszkiewicz PJ, et al. Targeted antibody-mediated depletion of murine CD19 CAR T cells permanently reverses B cell aplasia. J Clin Invest. 2016;126(11):4262–4272. doi: 10.1172/JCI84813. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 47.Juillerat A, et al. Modulation of chimeric antigen receptor surface expression by a small molecule switch. BMC Biotechnol. 2019;19(1):44. doi: 10.1186/s12896-019-0537-3. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 48.Mestermann K, et al. The tyrosine kinase inhibitor dasatinib acts as a pharmacologic on/off switch for CAR T cells. Sci Transl Med. 2019;11(499):eaau5907. doi: 10.1126/scitranslmed.aau5907. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 49.Fraietta JA, et al. Ibrutinib enhances chimeric antigen receptor T-cell engraftment and efficacy in leukemia. Blood. 2016;127(9):1117–1127. doi: 10.1182/blood-2015-11-679134. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 50.Gauthier J, et al. Feasibility and efficacy of CD19-targeted CAR T cells with concurrent ibrutinib for CLL after ibrutinib failure. Blood. 2020;135(19):1650–1660. doi: 10.1182/blood.2019002936. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 51.Jain MD, et al. Characteristics and outcomes of patients receiving bridging therapy while awaiting manufacture of standard of care axicabtagene ciloleucel CD19 chimeric antigen receptor (CAR) T-cell therapy for relapsed/refractory large B-cell lymphoma: results from the US lymphoma CAR-T consortium. Blood. 2019;134(Suppl_1):245 [Google Scholar]
  • 52.Torikai H, et al. A foundation for universal T-cell based immunotherapy: T cells engineered to express a CD19-specific chimeric-antigen-receptor and eliminate expression of endogenous TCR. Blood. 2012;119(24):5697–5705. doi: 10.1182/blood-2012-01-405365. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 53.Liu X, et al. CRISPR-Cas9-mediated multiplex gene editing in CAR-T cells. Cell Res. 2017;27(1):154–157. doi: 10.1038/cr.2016.142. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 54.Ren J, et al. Multiplex genome editing to generate universal CAR T cells resistant to PD1 inhibition. Clin Cancer Res. 2017;23(9):2255–2266. doi: 10.1158/1078-0432.CCR-16-1300. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 55.Mehta RS, Rezvani K. Chimeric antigen receptor expressing natural killer cells for the immunotherapy of cancer. Front Immunol. 2018;9:283. doi: 10.3389/fimmu.2018.00283. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 56.Liu E, et al. Use of CAR-transduced natural killer cells in CD19-positive lymphoid tumors. N Engl J Med. 2020;382(6):545–553. doi: 10.1056/NEJMoa1910607. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 57.Yu H, et al. Repeated loss of target surface antigen after immunotherapy in primary mediastinal large B cell lymphoma. Am J Hematol. 2017;92(1):E11–E13. doi: 10.1002/ajh.24594. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 58.Maude SL, et al. Chimeric antigen receptor T cells for sustained remissions in leukemia. N Engl J Med. 2014;371(16):1507–1517. doi: 10.1056/NEJMoa1407222. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 59.Sotillo E, et al. Convergence of acquired mutations and alternative splicing of CD19 enables resistance to CART-19 immunotherapy. Cancer Discovery. 2015;5(12):1282–1295. doi: 10.1158/2159-8290.CD-15-1020. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 60.Hamieh M, et al. CAR T cell trogocytosis and cooperative killing regulate tumour antigen escape. Nature. 2019;568(7750):112–116. doi: 10.1038/s41586-019-1054-1. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 61.Majzner RG, Mackall CL. Tumor antigen escape from CAR T-cell therapy. Cancer Discov. 2018;8(10):1219–1226. doi: 10.1158/2159-8290.CD-18-0442. [DOI] [PubMed] [Google Scholar]
  • 62.Zah E, et al. T Cells expressing CD19/CD20 bispecific chimeric antigen receptors prevent antigen escape by malignant B cells. Cancer Immunology Research. 2016;4(6):498–508. doi: 10.1158/2326-6066.CIR-15-0231. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 63.Qin H, et al. Preclinical development of bivalent chimeric antigen receptors targeting both CD19 and CD22. Mol Ther Oncolytics. 2018;11:127–137. doi: 10.1016/j.omto.2018.10.006. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 64.Pan J, et al. Sequential CD19-22 CAR T therapy induces sustained remission in children with r/r B-ALL. Blood. 2020;135(5):387–391. doi: 10.1182/blood.2019003293. [DOI] [PubMed] [Google Scholar]
  • 65.Jen EY, et al. FDA approval: blinatumomab for patients with B-cell precursor acute lymphoblastic leukemia in morphologic remission with minimal residual disease. Clin Cancer Res. 2019;25(2):473–477. doi: 10.1158/1078-0432.CCR-18-2337. [DOI] [PubMed] [Google Scholar]
  • 66.Choi BD, et al. CAR-T cells secreting BiTEs circumvent antigen escape without detectable toxicity. Nat Biotechnol. 2019;37(9):1049–1058. doi: 10.1038/s41587-019-0192-1. [DOI] [PubMed] [Google Scholar]
  • 67.Morgan RA, et al. Case report of a serious adverse event following the administration of T cells transduced with a chimeric antigen receptor recognizing ERBB2. Mol Ther. 2010;18(4):843–851. doi: 10.1038/mt.2010.24. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 68.Richman SA, et al. High-affinity GD2-specific CAR T cells induce fatal encephalitis in a preclinical neuroblastoma model. Cancer Immunol Res. 2018;6(1):36–46. doi: 10.1158/2326-6066.CIR-17-0211. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 69.Min IM, et al. CAR T therapy targeting ICAM-1 eliminates advanced human thyroid tumors. Clin Cancer Res. 2017;23(24):7569–7583. doi: 10.1158/1078-0432.CCR-17-2008. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 70.Kloss CC, et al. Combinatorial antigen recognition with balanced signaling promotes selective tumor eradication by engineered T cells. Nat Biotechnol. 2013;31(1):71–75. doi: 10.1038/nbt.2459. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 71.Roybal KT, et al. Engineering T cells with customized therapeutic response programs using synthetic notch receptors. Cell. 2016;167(2):419–432.e16. doi: 10.1016/j.cell.2016.09.011. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 72.Roybal KT, et al. Precision tumor recognition by t cells with combinatorial antigen-sensing circuits. Cell. 2016;164(4):770–779. doi: 10.1016/j.cell.2016.01.011. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 73.Sharifzadeh Z, et al. Genetically engineered T cells bearing chimeric nanoconstructed receptors harboring TAG-72-specific camelid single domain antibodies as targeting agents. Cancer Lett. 2013;334(2):237–244. doi: 10.1016/j.canlet.2012.08.010. [DOI] [PubMed] [Google Scholar]
  • 74.Siegler E, et al. Designed ankyrin repeat proteins as Her2 targeting domains in chimeric antigen receptor-engineered T cells. Hum Gene Ther. 2017;28(9):726–736. doi: 10.1089/hum.2017.021. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 75.Hammill JA, et al. Designed ankyrin repeat proteins are effective targeting elements for chimeric antigen receptors. J Immunother Cancer. 2015;3:55. doi: 10.1186/s40425-015-0099-4. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 76.He X, et al. Bispecific and split CAR T cells targeting CD13 and TIM3 eradicate acute myeloid leukemia. Blood. 2020;135(10):713–723. doi: 10.1182/blood.2019002779. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 77.Qin H, et al. Chimeric antigen receptors incorporating D domains targeting CD123 direct potent mono- and bi-specific antitumor activity of T cells. Mol Ther. 2019;27(7):1262–1274. doi: 10.1016/j.ymthe.2019.04.010. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 78.Priceman SJ, et al. Regional delivery of chimeric antigen receptor-engineered T cells effectively targets HER2 + breast cancer metastasis to the brain. Clinical Cancer Research. 2018;24(1):95–105. doi: 10.1158/1078-0432.CCR-17-2041. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 79.Tchou J, et al. Safety and efficacy of intratumoral injections of chimeric antigen receptor (CAR) T cells in metastatic breast cancer. Cancer Immunol Res. 2017;5(12):1152–1161. doi: 10.1158/2326-6066.CIR-17-0189. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 80.Katz SC, et al. Regional CAR-T cell infusions for peritoneal carcinomatosis are superior to systemic delivery. Cancer Gene Ther. 2016;23(5):142–148. doi: 10.1038/cgt.2016.14. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 81.van Schalkwyk MCI, et al. Design of a phase I clinical trial to evaluate intratumoral delivery of ErbB-targeted chimeric antigen receptor T-cells in locally advanced or recurrent head and neck cancer. Human Gene Therapy Clinical Development. 2013;24(3):134–142. doi: 10.1089/humc.2013.144. [DOI] [PubMed] [Google Scholar]
  • 82.Katz SC, et al. Phase I hepatic immunotherapy for metastases study of intra-arterial chimeric antigen receptor-modified T-cell therapy for CEA+ liver metastases. Clin Cancer Res. 2015;21(14):3149–3159. doi: 10.1158/1078-0432.CCR-14-1421. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 83.Adusumilli PS, et al. Regional delivery of mesothelin-targeted CAR T cell therapy generates potent and long-lasting CD4-dependent tumor immunity. Sci Transl Med. 2014;6(261):261ra151. doi: 10.1126/scitranslmed.3010162. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 84.Nagarsheth N, et al. Chemokines in the cancer microenvironment and their relevance in cancer immunotherapy. Nat Rev Immunol. 2017;17(9):559–572. doi: 10.1038/nri.2017.49. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 85.Craddock JA, et al. Enhanced tumor trafficking of GD2 chimeric antigen receptor T cells by expression of the chemokine receptor CCR2b. J Immunother. 2010;33(8):780–788. doi: 10.1097/CJI.0b013e3181ee6675. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 86.Moon EK, et al. Expression of a functional CCR2 receptor enhances tumor localization and tumor eradication by retargeted human T cells expressing a mesothelin-specific chimeric antibody receptor. Clin Cancer Res. 2011;17(14):4719–4730. doi: 10.1158/1078-0432.CCR-11-0351. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 87.Whilding LM, et al. CAR T-cells targeting the integrin αvβ6 and co-expressing the chemokine receptor CXCR2 demonstrate enhanced homing and efficacy against several solid malignancies. Cancers (Basel) 2019;11(5):674. doi: 10.3390/cancers11050674. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 88.Caruana I, et al. Heparanase promotes tumor infiltration and antitumor activity of CAR-redirected T lymphocytes. Nat Med. 2015;21(5):524–529. doi: 10.1038/nm.3833. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 89.Adachi K, et al. IL-7 and CCL19 expression in CAR-T cells improves immune cell infiltration and CAR-T cell survival in the tumor. Nat Biotechnol. 2018;36(4):346–351. doi: 10.1038/nbt.4086. [DOI] [PubMed] [Google Scholar]
  • 90.Xie YJ, et al. Nanobody-based CAR T cells that target the tumor microenvironment inhibit the growth of solid tumors in immunocompetent mice. Proc Natl Acad Sci U S A. 2019;116(16):7624–7631. doi: 10.1073/pnas.1817147116. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 91.Ma S, et al. Current progress in CAR-T cell therapy for solid tumors. Int J Biol Sci. 2019;15(12):2548–2560. doi: 10.7150/ijbs.34213. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 92.Rafiq S, et al. Targeted delivery of a PD-1-blocking scFv by CAR-T cells enhances anti-tumor efficacy in vivo. Nat Biotechnol. 2018;36(9):847–856. doi: 10.1038/nbt.4195. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 93.Suarez ER, et al. Chimeric antigen receptor T cells secreting anti-PD-L1 antibodies more effectively regress renal cell carcinoma in a humanized mouse model. Oncotarget. 2016;7(23):34341–34355. doi: 10.18632/oncotarget.9114. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 94.Cherkassky L, et al. Human CAR T cells with cell-intrinsic PD-1 checkpoint blockade resist tumor-mediated inhibition. J Clin Invest. 2016;126(8):3130–3144. doi: 10.1172/JCI83092. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 95.Hu B, et al. Nucleofection with plasmid DNA for CRISPR/Cas9-mediated inactivation of programmed cell death protein 1 in CD133-specific CAR T cells. Hum Gene Ther. 2018;30(4):446–458. doi: 10.1089/hum.2017.234. [DOI] [PubMed] [Google Scholar]
  • 96.Rupp LJ, et al. CRISPR/Cas9-mediated PD-1 disruption enhances anti-tumor efficacy of human chimeric antigen receptor T cells. Sci Rep. 2017;7(1):737. doi: 10.1038/s41598-017-00462-8. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 97.Hu W, et al. CRISPR/Cas9-mediated PD-1 disruption enhances human mesothelin-targeted CAR T cell effector functions. Cancer Immunol Immunother. 2019;68(3):365–377. doi: 10.1007/s00262-018-2281-2. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 98.Fu W, et al. CAR exosomes derived from effector CAR-T cells have potent antitumour effects and low toxicity. Nat Commun. 2019;10(1):4355. doi: 10.1038/s41467-019-12321-3. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 99.Jacobson CA, et al. Phase 1/2 primary analysis of ZUMA-6: Axicabtagene ciloleucel (Axi-Cel) in combination With atezolizumab (Atezo) for the treatment of patients (Pts) with refractory diffuse large B cell lymphoma (DLBCL) Cancer Res. 2020;80(16):Abstract nr CT055 [Google Scholar]
  • 100.Heczey A, et al. CAR T cells administered in combination with lymphodepletion and PD-1 inhibition to patients with neuroblastoma. Mol Ther. 2017;25(9):2214–2224. doi: 10.1016/j.ymthe.2017.05.012. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 101. Adusumilli PS, et al. AACR; 2019.Abstract CT036: A phase I clinical trial of malignant pleural disease treated with regionally delivered autologous mesothelin-targeted CAR T cells: Safety and efficacy. Presented at: AACR Annual Meeting 2019; March 29–April 3, 2019; Atlanta, GA. [DOI] [Google Scholar]
  • 102.Koneru M, et al. IL-12 secreting tumor-targeted chimeric antigen receptor T cells eradicate ovarian tumors in vivo. Oncoimmunology. 2015;4(3):e994446. doi: 10.4161/2162402X.2014.994446. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 103.Liu Y, et al. Armored inducible expression of IL-12 enhances antitumor activity of glypican-3-targeted chimeric antigen receptor-engineered T cells in hepatocellular carcinoma. J Immunol. 2019;203(1):198–207. doi: 10.4049/jimmunol.1800033. [DOI] [PubMed] [Google Scholar]
  • 104.Yeku OO, et al. Armored CAR T cells enhance antitumor efficacy and overcome the tumor microenvironment. Sci Rep. 2017;7(1):10541. doi: 10.1038/s41598-017-10940-8. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 105.Pegram HJ, et al. Tumor-targeted T cells modified to secrete IL-12 eradicate systemic tumors without need for prior conditioning. Blood. 2012;119(18):4133–4141. doi: 10.1182/blood-2011-12-400044. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 106.Kueberuwa G, et al. CD19 CAR T cells expressing IL-12 eradicate lymphoma in fully lymphoreplete mice through induction of host immunity. Mol Ther Oncolytics. 2017;8:41–51. doi: 10.1016/j.omto.2017.12.003. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 107.Chen Y, et al. Eradication of neuroblastoma by T cells redirected with an optimized GD2-specific chimeric antigen receptor and interleukin-15. Clin Cancer Res. 2019;25(9):2915–2924. doi: 10.1158/1078-0432.CCR-18-1811. [DOI] [PubMed] [Google Scholar]
  • 108.Krenciute G, et al. Transgenic expression of IL15 improves antiglioma activity of IL13Rα2-CAR T cells but results in antigen loss variants. Cancer Immunol Res. 2017;5(7):571–581. doi: 10.1158/2326-6066.CIR-16-0376. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 109.Chmielewski M, Abken H. CAR T cells releasing IL-18 convert to T-Bethigh FoxO1low effectors that exhibit augmented activity against advanced solid tumors. Cell Rep. 2017;21(11):3205–3219. doi: 10.1016/j.celrep.2017.11.063. [DOI] [PubMed] [Google Scholar]
  • 110.Hu B, et al. Augmentation of antitumor immunity by human and mouse CAR T cells secreting IL-18. Cell Rep. 2017;20(13):3025–3033. doi: 10.1016/j.celrep.2017.09.002. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 111.Tanaka A, Sakaguchi S. Regulatory T cells in cancer immunotherapy. Cell Res. 2017;27(1):109–118. doi: 10.1038/cr.2016.151. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 112.Ostrand-Rosenberg S, Fenselau C. Myeloid-derived suppressor cells: immune-suppressive cells that impair antitumor immunity and are sculpted by their environment. J Immunol. 2018;200(2):422–431. doi: 10.4049/jimmunol.1701019. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 113.Burga RA, et al. Liver myeloid-derived suppressor cells expand in response to liver metastases in mice and inhibit the anti-tumor efficacy of anti-CEA CAR-T. Cancer Immunol Immunother. 2015;64(7):817–829. doi: 10.1007/s00262-015-1692-6. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 114.Long AH, et al. Reduction of MDSCs with all-trans retinoic acid improves CAR therapy efficacy for sarcomas. Cancer Immunol Res. 2016;4(10):869–880. doi: 10.1158/2326-6066.CIR-15-0230. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 115.Mardiana S, et al. A multifunctional role for adjuvant anti-4-1BB therapy in augmenting antitumor response by chimeric antigen receptor T cells. Cancer Res. 2017;77(6):1296–1309. doi: 10.1158/0008-5472.CAN-16-1831. [DOI] [PubMed] [Google Scholar]
  • 116.Klichinsky M, et al. Human chimeric antigen receptor macrophages for cancer immunotherapy. Nat Biotechnol. 2020;38(8):947–953. doi: 10.1038/s41587-020-0462-y. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 117.Gattinoni L, 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(7):907–912. doi: 10.1084/jem.20050732. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 118.Suryadevara CM, et al. Preventing Lck activation in CAR T cells confers treg resistance but requires 4-1BB signaling for them to persist and treat solid tumors in nonlymphodepleted hosts. Clin Cancer Res. 2019;25(1):358–368. doi: 10.1158/1078-0432.CCR-18-1211. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 119.Chinnasamy D, et al. Local delivery of interleukin-12 using T cells targeting VEGF receptor-2 eradicates multiple vascularized tumors in mice. Clin Cancer Res. 2012;18(6):1672–1683. doi: 10.1158/1078-0432.CCR-11-3050. [DOI] [PMC free article] [PubMed] [Google Scholar]

Articles from The Journal of Clinical Investigation are provided here courtesy of American Society for Clinical Investigation

RESOURCES