Abstract
Chimeric antigen receptor T (CAR-T) cell therapy is a promising and precise targeted therapy for cancer that has demonstrated notable potential in clinical applications. However, severe adverse effects limit the clinical application of this therapy and are mainly caused by uncontrollable activation of CAR-T cells, including excessive immune response activation due to unregulated CAR-T cell action time, as well as toxicity resulting from improper spatial localization. Therefore, to enhance controllability and safety, a control module for CAR-T cells is proposed. Synthetic biology based on genetic engineering techniques is being used to construct artificial cells or organisms for specific purposes. This approach has been explored in recent years as a means of achieving controllability in CAR-T cell therapy. In this review, we summarize the recent advances in synthetic biology methods used to address the major adverse effects of CAR-T cell therapy in both the temporal and spatial dimensions.
Synthetic biology contributes to the realization of intelligent CAR-T cell therapies with enhanced safety and controllability.
INTRODUCTION
Adoptive T cell therapy is a prominent therapeutic method that demands precise T cell editing and signal regulation. This process involves ex vivo expansion of tumor-specific T cells followed by their reintroduction into patients to swiftly restore host immunity (1). Recent clinical trials have demonstrated that engineered T cells are highly active tumor effector cells. Among these, chimeric antigen receptor T (CAR-T) cell therapy has shown promising outcomes in the management of diverse malignancies, particularly hematological cancers. The activation intensity of CAR-T cells plays a crucial role in determining their effectiveness and is closely linked to CAR design. The initial generation of CAR exhibited suboptimal activation due to the absence of a second signal in the intracellular segment, which is vital for T cell activation. Subsequent modifications involved the incorporation of one or more costimulatory molecules, such as CD28 or 4-1BB, which substantially promote activation. However, the adverse effects were also more pronounced.
The safety concerns associated with CAR-T cells can be divided into the following three types: cytokine release syndrome (CRS), immune effector cell–associated neurotoxicity syndrome (ICANS), and on-target off-tumor (OTOT) toxicity. These adverse effects primarily result from unregulated activation and can be attributed to two factors, excessive activation of the immune system due to the uncontrolled timing and extent of CAR-T cell effects and toxicity to normal cells due to uncontrolled off-tumor phenomenon in space. Furthermore, toxicity is also associated with treatment-related factors, such as CAR-T cell dosage and drug conditioning chemotherapy, as well as factors related to patient heterogeneity, including tumor burden, the presence of underlying disease, and the levels of relevant biomarkers. At present, the use of the interleukin-6 (IL-6) receptor antagonist tocilizumab (2) and the IL-1 receptor antagonist anakinra (3) is the main clinical approach used to alleviate CRS and ICANS. However, while these medications inhibit the overactivation of CAR-T cells, they also suppress the normal immune function of the host’s endogenous T cells, potentially leading to diminished immune capability and increased susceptibility to infections. For OTOT toxicity, the ideal solution is to select strict tumor-specific antigens (TSAs) as targets in CAR design. However, the identification of such TSAs poses challenges, and most of the targets that are currently used in clinical practice still carry the risk of OTOT toxicity. Consequently, there is a pressing need for approaches to enhance the controllability of CAR-T cells while limiting their cytotoxic effects.
Synthetic biology is a burgeoning discipline that uses engineering principles to reengineer and modify natural biological systems while constructing novel standardized biological components and systems. Characterized by its systematic, modular, and standardized engineering features, synthetic biology offers theoretical guidance and technical support for engineered cell therapies. Given the intricate nature and controllability of the T cell activation signaling pathway, T cells and associated signaling systems serve as ideal platforms for synthetic engineering (4). Consequently, CAR-T cells represent a primary application of synthetic biology within the realm of immunology. In this review, we summarize the latest advances in the application of synthetic biology methods for controlling CAR-T cells both temporally and spatially to reduce adverse effects. In addition, we address the persisting challenges and outline future prospects in this field.
THE TOXICITY OF CAR-T CELL THERAPY
The common cause of adverse effects in patients receiving CAR-T cell therapy is uncontrolled activation. The effects can be divided into two groups based on temporal and spatial dimensions (Fig. 1).
Fig. 1. Overview of the toxicity of CAR-T cell therapy.
In the temporal dimension, CAR-T cells are overactivated to produce excessive amounts of cytokines, resulting in CRS. Subsequently, immune cells and cytokines from the periphery infiltrate the central nervous system, leading to ICANS. In the spatial dimension, CAR-T cells may identify target antigens on normal cells, resulting in OTOT toxicity. Figure created with BioRender.com.
In the temporal dimension, prolonged or excessive CAR-T cell activity can lead to an excessive release of cytokines and overactivation of the immune system. CRS is the prevailing toxic reaction (5, 6), and the incidence of grade 3 or above CRS has reached 30% in some clinical trials (Table 1). CRS arises because of high-level immune activation and is characterized by elevated levels of interferon-γ (IFN-γ), granulocyte-macrophage colony-stimulating factor (GM-CSF), IL-2, IL-8, IL-5, IL-6, IL-10, or tumor necrosis factor–α (TNF-α) (7). Symptoms of CRS include fever, hypoxia, terminal organ dysfunction, cytopenia, hypotension, coagulation dysfunction, and hemophagocytic lymphohistiocytosis (8). Increased cytokine levels in the cerebrospinal fluid and destruction of the blood-brain barrier (BBB) are the causes of ICANS (9, 10). The reported incidence of ICANS is approximately 0 to 73%, and ICANS occurs simultaneously with or after CRS in most patients (Table 1) (11). Symptoms of ICANS include encephalopathy, cognitive deficits, speech disorders, seizures, and brain edema. Early alterations in cytokine profiles (IFN-γ, soluble gp130, IL-6, and soluble IL-6 receptor) or serum biochemical marker levels (ferritin and C-reactive protein) can be used to evaluate CRS and ICANS severity (12). Teachey et al. (13) developed a model to predict the occurrence of CRS by monitoring the levels of IFN-γ, IL-1, and MIP72α. ICANS biomarkers mainly include proinflammatory activation markers (IL-1 and IL-6), pro-proliferative markers (IL-15 and GM-CSF), immunomodulators (IL-10 and IFN-γ), and baseline systemic factors (14). Interventions that can reduce or completely inhibit CAR-T cell function when relevant biomarkers are detected are needed to avoid cytokine storms.
Table 1. Incidences of CRS and ICANS of CAR-T cell therapy overviewing most recent research (2022–2023).
GPRC5D, G protein–coupled receptor class C group 5 member D; BCMA, B cell maturation antigen; CD19, cluster of differentiation 19; CD22, cluster of differentiation 22; CD7, cluster of differentiation 7; CD20, cluster of differentiation 20; PSMA, prostate-specific membrane antigen; CLDN18, claudin-18; CLDN6, claudin 6; MET, mesenchymal epithelial transition factor; EpCAM, epithelial cell adhesion molecule.
| Disease | Target | Incidence of CRS (%) | Incidence of ≥grade 3 CRS (%) | Incidence of ICANS (%) | Incidence of ≥grade 3 ICANS (%) | Clinical trial identifier | Refs. |
|---|---|---|---|---|---|---|---|
| Multiple myeloma | GPRC5D | 100 | 0 | 0 | 0 | NCT05016778 | (115) |
| GPRC5D | 76 | 0 | 9 | 3 | ChiCTR2100048888 | (116) | |
| BCMA | 88 | 5 | 15 | 3 | NCT03651128 | (117) | |
| BCMA | 56 | 2 | 14 | 0 | NCT04093596 | (118) | |
| BCMA | 94 | 10 | 3 | 0 | NCT03815383NCT03751293NCT04295018NCT04322292 | (119) | |
| BCMA | 92 | 10 | 1 | 0 | NCT03090659 | (120) | |
| BCMA | 35 | 6 | 0 | 0 | NCT03093168 | (121) | |
| BCMA | 100 | 8 | 17 | 8 | NCT04155749 | (122) | |
| BCMA | 76 | 1 | 5 | 0 | NCT04181827 | (123) | |
| BCMA | 90 | 0 | 0 | 0 | NCT04720313 | (124) | |
| BCMA | 94 | 28 | 39 | 11 | NCT03502577 | (125) | |
| BCMA | 84 | 4 | 18 | 4 | NCT03361748 | (126) | |
| BCMA | 76 | 7 | 37 | 2 | NCT02658929 | (127) | |
| CD19/CD22 | 27 | 0 | 7 | 0 | NCT03919526 | (128) | |
| CD19/CD22 | 50 | 15 | 5 | 5 | NCT03448393 | (129) | |
| CD19 | 96 | 24 | 28 | 28 | NCT03825718 | (130) | |
| CD19 | 50 | 17 | 25 | 8 | NCT03573700 | (131) | |
| Acute lymphoblastic leukemia with central nervous system lymphoma | CD19 | 90 | 19 | 38 | 23 | NCT02782351 | (132) |
| Primary central nervous system lymphoma | CD19 | 58 | 0 | 50 | 8 | NCT02445248 | (133) |
| Acute lymphoblastic leukemia | CD7 | 83 | 0 | 0 | 0 | NCT04538599 | (134) |
| CD7 | 100 | 0 | 67 | 0 | ISRCTN15323014 | (135) | |
| CD19/CD22 | 88 | 28 | 21 | 4 | ChiCTR2000032211 | (3) | |
| CD19/CD22 | 99 | 19 | 37 | 5 | NCT04340154 | (136) | |
| Large B cell lymphoma | CD19 | 38 | 2 | 31 | 5 | NCT03483103 | (137) |
| CD19 | 50 | 1 | 12 | 4 | NCT03575351 | (138) | |
| CD19 | 50 | 0 | 10 | 0 | NCT03484702 | (139) | |
| CD19 | 100 | 8 | 73 | 23 | NCT03761056 | (140) | |
| CD19 | 74 | 6 | 19 | 10 | NCT04148430 | (3) | |
| CD19 | 85 | 10 | 50 | 25 | NCT03954106 | (141) | |
| CD19 | 59 | 8 | 8 | 0 | NCT03630159 | (142) | |
| CD19 | 48 | 5 | 20 | 3 | NCT04089215 | (143) | |
| B cell non-Hodgkin lymphoma | CD19/CD20 | 70 | 10 | 17 | 2 | NCT03097770 | (144) |
| CD19 | 18 | 5 | 23 | 0 | NCT03344367 | (145) | |
| B cell malignancies | CD19 | 21 | 0 | 0 | 0 | NCT02807883 | (146) |
| CD19 | 43 | 0 | 29 | 0 | NCT04206943 | (147) | |
| T cell malignancies | CD7 | 95 | 5 | 10 | 0 | NCT04572308 | (148) |
| CD7 | 100 | 13 | 0 | 0 | NCT04004637 | (149) | |
| Neuromyelitis optica spectrum disorder | BCMA | 100 | 0 | 0 | 0 | NCT04561557 | (150) |
| Gastric cancer | CLDN18 | 95 | 0 | 0 | 0 | NCT03874897 | (151) |
| Metastatic castration-resistant prostate cancer | PSMA | 60 | 30 | 10 | 10 | NCT03089203 | (152) |
| Solid tumors | CLDN6 | 46 | 5 | 5 | 0 | NCT04503278 | (153) |
| MET | 14 | 0 | 0 | 0 | NCT03060356 | (154) | |
| Epithelial tumors | EpCAM | 0 | 0 | 17 | 0 | NCT02915445 | (155) |
| Myasthenia gravis | BCMA | 0 | 0 | 0 | 0 | NCT04146051 | (156) |
In the spatial dimension, CAR-T cells may be localized and uncontrollably activated toward normal cells that express target antigens, resulting in OTOT toxicity. The ideal antigen should be expressed only on tumor cells (called TSAs, such as neoantigens), but few TSAs are currently available (15). Thus, tumor-associated antigens (TAAs), including human epithelial growth factor receptor-2 (HER2), epidermal growth factor receptor (EGFR), B7 homolog 3 (B7-H3), carbonic anhydrase IX (CAIX), mesothelin, and ganglioside 2 (GD2), are chosen as targets; however, all of these antigens exhibit a certain degree of expression in normal tissues (16, 17). Although the amount of TAAs expressed in normal cells is substantially lower than that in tumor cells, CAR-T cells can respond to low-expression antigens due to their high sensitivity. When exposed to antigens, infused CAR-T cells can undergo activation and proliferation by several orders of magnitude, potentially leading to severe toxicity. Table 2 presents a comprehensive list of representative clinical cases of OTOT toxicity during treatment. For instance, CAIX-targeted CAR-T cells recognize normal epithelial cells of the inner wall of the bile duct, thereby causing discrete cholangitis (18, 19); HER2-targeted CAR-T cells can lead to OTOT toxicity, such as mild skin itching and severe upper gastrointestinal bleeding (20); and EGFR-targeted CAR-T cells can cause mucosal and skin side effects (21–23). Therefore, the pressing challenge lies in achieving precise spatial localization of CAR-T cells within tumor tissues and mitigating adverse effects.
Table 2. Typical cases of OTOT toxicity of CAR-T cell therapy overviewing recent research.
HER2, human epidermal growth factor receptor 2; CEACAM5, carcinoembryonic antigen cell adhesion molecule 5 gene; CAIX, carbonic anhydrase IX; EGFR, epidermal growth factor receptor; CD133, cluster of differentiation 133.
| Disease | Target | Off-tumor site | Result | Clinical trials identifier | Refs. |
|---|---|---|---|---|---|
| Colon cancer | HER2 | Lungs and liver | High doses of CAR-T cells targeting HER2 on normal lung cells cause fatal cytokine storms | NCI-09-C-0041 | (157) |
| Biliary tract cancers and pancreatic cancers | HER2 | Gastrointestinal mucosa | Upper gastrointestinal hemorrhage | NCT01935843 | (20) |
| Gastrointestinal malignancies | CEACAM5 | Lungs | Respiratory toxicity | NCT01212887 | (158) |
| Renal cell carcinoma | CAIX | Bile duct epithelium | Liver toxicity | DDHK97-29/P00.0040C | (18, 19, 159) |
| Biliary tract cancer | EGFR | Epidermis | Skin rashes | NCT01869166 | (22) |
| Cholangiocarcinoma | CD133 | Epithelium and vascular endothelium | Severe dermatologic, oral mucosal, and gastrointestinal toxicities | NCT02541370 | (160) |
| B cell lymphoma and acute lymphoblastic leukemia | CD19 | Normal B cells | B cell depletion, hypogammaglobulinemia | NCT00924326 | (161) |
| Gastric cancer | CLDN18 | Gastric mucosa | Mucosal erosion | NCT03874897 | (151) |
THE SECURITY GUARANTEES OF CAR-T CELLS IN THE TEMPORAL DIMENSION
T cell immunity in the human body can be regulated by activation or inactivation through various mechanisms, thereby enabling the balance of activity and safety. Consequently, the incorporation of a regulatory module into CAR-T cells could be used to achieve regulated activation, substantially enhancing the safety of this therapy while maintaining its effectiveness. The characteristics and advantages of synthetic biology include the combination of biology and engineering, which allows for the modularization, hierarchy, standardization, and quantification of biological functions. By using the logic-gating principles of electric circuits to combine standardized regulatory elements such as promoters, repressors, enhancers, and regulated genes, gene circuits can be created. The integration of these genetic circuits into T cells or the modification of chassis T cells can be used to effectively address safety concerns (Fig. 2).
Fig. 2. Synthetic biology methods can effectively improve the safety and controllability of CAR-T cells.
In the temporal dimension, the synthetic biology approaches for guaranteeing safety include killing switches, adapter switches, small-molecule drug switches, and chassis cell modification. In the spatial dimension, methods include Boolean logic gates (AND gates, NAND gates) constructed from synthetic receptors, redirected migration of T cells, and targeting the tumor microenvironment. Dual control of the temporal and spatial dimensions can be achieved by light-operated switches and ultrasound-operated switches. Figure created with BioRender.com.
Killing switches
The activation of killing switches is induced by specific small molecules, which initiate the programmed cell death pathway that results in irreversible termination of the immune response. Incorporating killing switches is among the straightforward and highly effective approaches for enhancing the controllability of CAR-T cells.
On the basis of their diverse underlying mechanisms, killing switches can be classified into three categories: inducible caspase-9 suicide gene system (iCas9) switches, ganciclovir-activated switches, and antibody-dependent cell-mediated cytotoxicity (ADCC) switches. Fusion proteins with modified human caspase-9 and human FK506-binding proteins (FKBPs) are the main components of iCas9 switches. Chemical inducers of dimerization (CIDs), such as AP1903/rimiducid or AP20187, can mediate FKBP12-F36V homodimerization, after which caspases send an apoptotic signal to the cell. iCas9 switches have shown high effectiveness and have been the subject of numerous clinical trials (Table 3). According to clinical data, CID can eliminate CAR-T cells in the peripheral blood and central nervous system, resulting in the prompt alleviation of adverse events such as graft-versus-host disease (GVHD) and CRS (24, 25). Furthermore, this control mechanism remains effective even when CAR-T cells undergo a sharp decrease after drug administration and subsequent reamplification over time (26). Ganciclovir-activated switches turn ganciclovir into a toxic metabolite using the herpes simplex virus-1 thymidine kinase (HSV-TK) protein, which leads to cell death. The administration of ganciclovir effectively eliminated CAR-T cells and suppressed their functionality in a xenograft model (27). Since HSV-TK is an exogenous virus-derived protein, the use of ganciclovir-activated switches may pose a safety concern regarding immunogenicity in contrast to iCas9 (28). Moreover, the activation of HSV-TK by ganciclovir in vitro requires a period of 3 days, which is considered relatively slow (29). HSV-TK can also be used as an enzyme-based reporter gene for positron emission tomography (PET) imaging (30), and it has been successfully used in a phase 1 clinical trial to visualize CAR-T cells within the bodies of seven glioblastoma patients (31). The combination of the HSV-TK reporter with synthetic Notch (synNotch) enables the imaging of CAR-T cells that specifically bind to their target antigens (32). A truncated protein such as a truncated human EGFR polypeptide (huEGFRt) that is recognized by monoclonal antibodies (mAbs) is coexpressed and helps make up ADCC switches. After the provision of mAbs, CAR-T cells can be eliminated by ADCC or complement-dependent cytotoxicity (CDC). JCAR017 is a CAR-T cell product containing huEGFRt that can be recognized by cetuximab and rendered ineffective; this product showed superior safety in the TRANSCEND NHL 001 trial (NCT02631044) (33). The ADCC switches often take longer to work than the killing switches discussed above. In addition, the ability of cetuximab to penetrate the BBB is highly restricted (34), which could restrict the ability of ADCC switches to alleviate CRS and ICANS. Moreover, lymphodepletion is administered to patients before CAR-T cell therapy in current clinical practice, aiming to reduce the circulating immune cell count and enhance the antitumor efficacy of CAR-T cell therapy. Therefore, this switch is not applicable in such circumstances (35).
Table 3. Examples of applying synthetic biology methods to enhance the safety of CAR-T cell therapy for spatiotemporal regulation.
| Dimension | Category | Type | Phase | Evidence of mitigating CRS/ICANS/OTOT toxicity | Refs. |
|---|---|---|---|---|---|
| Security guarantee in the temporal dimension | Killing switches | iCas9 switch | Phase 1/2 NCT03373097 | Following the administration of rimiducid, a precipitous decline in the peripheral blood CAR-T cell count was observed, and this reduced level was sustained after the subsequent dose. After 6 weeks, CAR-T cells began to reamplified and could still be controlled by rimiducid. | (26) |
| Phase 1/2 NCT03016377 | After administering rimiducid, there was a notable improvement in ICANS symptoms, coupled with the removal of >60% of the CAR-T cells from the circulation within 4 hours and >90% within 24 hours. | (162) | |||
| Phase 1 NCT01494103 | Approximately 85–95% of CD3+CD19+ T cells in the bloodstream and over 90% in the cerebrospinal fluid were eradicated subsequent to the administration of CID. GVHD-related CRS were relieved within 2 hours. | (24) | |||
| Phase 1 NCT03170141 | The safety of CAR-T cells is considered acceptable. iCas9 switch was armored but not used. | (163) | |||
| Phase 1 NCT04196413 | The safety of CAR-T cells is considered acceptable. iCas9 switch was armored but not used. | (164) | |||
| Phase 1 NCT01822652 | The safety of CAR-T cells is considered acceptable. iCas9 switch was armored but not used. | (165) | |||
| Phase 1/2 NCT02414269 | The safety of CAR-T cells is considered acceptable. iCas9 switch was armored but not used. | (166) | |||
| ADCC switch | Phase 1 NCT02631044 | The safety of CAR-T cells is considered acceptable. ADCC switches were armored but not used. | (33) | ||
| Phase 1 NCT02311621 | The safety of CAR-T cells is considered acceptable. ADCC switches were armored but not used. | (167) | |||
| Phase 1 NCT03618381 | The results of the clinical trial have not been disclosed yet. | / | |||
| Phase 1 NCT02498912 | The results of the clinical trial have not been disclosed yet. | (168) | |||
| Ganciclovir activated switch | Phase 1BB-IND 9149 | An immune response against CAR-T cells emerged, limiting the efficacy and persistence of subsequent infusions. | (169) | ||
| Phase 1 NCT00730613 | The safety of CAR-T cells is considered acceptable. Hy-TK killing switch was armored but not used. | (170) | |||
| Phase 1/2 NCT04097301 terminated | The clinical trial has been terminated. HSV-TK killing switch was armored but not used. | (171) | |||
| Preclinical study | The administration of ganciclovir can effectively eliminate CAR-T cells in vivo and suppresses their functionality in mice. | (27) | |||
| Adapter switches | FITC-folate adapter switch | Phase 1 NCT05312411 | The results of the clinical trial have not been disclosed yet. | / | |
| Preclinical study | It can quickly terminate CRS-like toxicity within 3 hours and preemptively prevent the occurrence of CRS-like toxicity. | (39) | |||
| CD123-specific targeting module | Phase 1 NCT04230265 | The clinical trial results were unpublished. TM123 UniCAR-T exhibited reversible toxicity in mice, unlike CD123 CAR-T with notable hematological toxicity. | (52) | ||
| Small-molecule switches | On switch | Phase 1/2 NCT02744287 Suspended | The CAR-T cells equipped with the iMC on switch exhibited rimiducid-dependent activity. Nevertheless, clinical trials were halted owing to pronounced adverse reactions. | (72) | |
| Preclinical study | The activity of CD19-DARIC T cells in vivo is completely dependent on rapamycin-mediated control. | (61) | |||
| Off switch | Preclinical study | Precise modulation of CAR expression levels using small molecules enables reversible control of CAR-T cell activity in vivo. | (77) | ||
| Chassis cell modification | Metabolic engineering modification | Preclinical study | Two weeks following administration of CAR-T cells in the absence of exogenous uridine, cell counts decreased substantially in vivo. | (83) | |
| Security guarantee in the spatial dimensions | Boolean logic gates constructed by synthetic receptors | AND gate (synNotch) | Phase 1 NCT05617755 | The findings from the clinical trial have not been published. However, in preclinical investigations, it has demonstrated robust antitumor efficacy and exhibited a high degree of specificity. | (172) |
| A AND NOT B gate (CAR + iCAR) | Phase 1/2 NCT05736731 | The findings from the clinical trial have not been published. Preclinical study showed that engineered T cells selectively kill HLA-A*02–negative cells but not HLA-A*02–expressing cells in vitro and in vivo. | (94) | ||
| Redirected migration of T cells | Cytokine-operated | Phase 1 NCT04153799 | The results of the clinical trial have not been disclosed. The in vitro preparation method for CAR-T cells incorporating CXCR5 appears to be feasible. | (173) | |
| Preclinical study | The T cell infiltration elicited by CXCR2-expressing T cell therapy exhibited a more than twofold increase compared to the control T cell therapy. | (98) | |||
| Orthogonal receptor-drug pair | Preclinical study | T lymphocytes can be specifically localized and last for at least 7 days in vivo. | (100) | ||
| Light-operated | Preclinical study | The infiltration of T lymphocytes at the tumor site was substantially enhanced upon local light stimulation. | (101) | ||
| Targeting tumor microenvironment | Trending toward growth-inhibitory acidic TME | Phase 1/2 NCT03393936 | No dose-limiting toxicities have been observed to date with no indications of OTOT toxicity attributed to either product. | (174) | |
| Dual oxygen-sensing switch | Preclinical study | A preclinical study had shown that this hypoxia-regulated CAR expression system substantially improves its antitumor specificity. | (104) | ||
| Dual security guarantee in the temporal and spatial dimensions | Light-operated switches | UV-sensitive photolyzable switch | Preclinical study | A preclinical study had shown that the activity of CAR-T cells is in a dose-dependent manner by the mediator and time-spatial controlled by UV. | (108) |
| Ultrasound-operated switches | Focused ultrasound switch | Preclinical study | The effectiveness of FUS-CAR-T cells to non–UV-irradiated tissues (expressing target antigens) was only 9.8%, while that of the standard CAR-T cells was 99.8%. | (112) |
The killing switch, which is a well-established synthetic biology approach, has been implemented in numerous clinical trials (NCT00710892, NCT03373097, NCT02631044, etc.). However, there are still critical issues that need to be addressed. First, the inducer (AP1903 or AP20187) for the iCas9 switches used in clinical trials has not been approved for sale. This limitation restricts the clinical application of the suicide gene system, making the selection of commercially available drugs a more practical choice. The RapaCaspase9 suicide gene uses the readily available marketed drug rapamycin and has demonstrated favorable outcomes in preclinical experiments, indicating promising potential for future clinical applications (36). Second, upon activation, these switches initiate an irreversible death mechanism, posing considerable challenges for both the high manufacturing costs and the prolonged persistence of CAR-T cells after treatment. In addition, suicide genes originating from nonhuman sequences may increase the immunogenicity risk of CAR-T cells, inducing both humoral and cellular immune responses (37). This intensifies therapeutic risks and the regulatory burden for post-administration patient monitoring. Compared to virus-derived HSV-TK, iCas9, which is of human origin, carries a reduced risk of immunogenicity. Clinical reports indicate that T cells harboring the iCas9 suicide gene in the human body persist for an extended period (38).
Adapter switches
The adapter molecule functions as a bridging molecule, binding one end to the universal CAR on CAR-T cells and the other end to the antigen on tumor cells. The “on” or “off” state and the degree of the effect are regulated by adjusting the presence and quantity of adapter molecules. Lee et al. (39) demonstrated that by regulating the concentration and dosing of the adapter molecule, CRS-like toxicity can be quickly terminated or prevented. Furthermore, adapter switches enable swift target switching, and the associated CAR-T cells demonstrate a high level of universality, thereby improving programmability and reducing therapeutic costs.
Biological orthogonal pairs are the foundation of adapter switches and provide the adapter molecule with a high degree of specificity for binding to CARs. The split, universal, and programmable (SUPRA) CAR serves as an exemplary adapter switch system. The zipFv functions as an adapter molecule that consists of a single-chain variable fragment (scFv) that specifically binds to tumor cells and a leucine zipper that interacts with the zipCAR. This enables simultaneous binding to both tumor cells and engineered T cells expressing zipCAR, which leads to the activation of cytotoxic functions (40). A major advantage of this switch is that when a higher-affinity zipFv is added, it can compete with the original zipFv, thereby converting the switch to off (41). Another classic example of a biomolecular orthogonal pair is the fluorescein isothiocyanate (FITC)–anti-FITC system, which achieves specific recognition of tumor antigens and anti-FITC CAR by coupling FITC with antibodies (42–46). The FITC-folate adapter switch is a relatively mature design and has undergone evaluation in a phase 1 trial for osteosarcoma (NCT05312411). In addition, the biotin-avidin system can specifically bind biotin-labeled antibodies to CARs equipped with avidin (47, 48). The PNE-mAb 52SR4 system links the peptide neoepitope (PNE), derived from the GCN4 peptide sequence, with a specific antibody. This enables the system to identify CARs that carry the mouse mAb 52SR4 in an scFv format (46, 49, 50). The bacterial toxin-antitoxin barnase-barstar system is a biomolecular orthogonal pair that can be applied in CAR-T cell therapy. The engineered chimeric receptor, which incorporates barstar, is designed to specifically bind to the tumor-targeted barnase (51).
Adapter switches demonstrate promising prospects for practical applications. The CD123-specific targeting module (TM123) is an adaptor that connects the universal CAR-T cell platform and CD123-positive leukemia cells (52). Meyer et al. developed a TM123 variant (TM123-4-1BBL) that can specifically bind to CD123-positive acute myeloid leukemia (AML) cell lines, universal CAR, and the 4-1BB costimulatory receptor, which can enhance persistence and effector function. This switch improves the controllability of the system, which may misrecognize normal hematopoietic progenitor cells and endothelial cells expressing CD123 (52, 53). The receptor tyrosine kinase–like orphan receptor 1 (ROR1) has up-regulated expression in bone marrow stromal cells after chemotherapy and is prominently expressed in the lungs. Consequently, conventional CAR-T cells targeting ROR1 have been found to induce lethal bone marrow failure and pulmonary toxicity in mice (54, 55). Switchable CAR-T (sCAR-T) systems provide a systematic and precisely controllable system for CAR-T cell targeting of ROR1 through the use of the GCN4 peptide–tagged (recognizing mouse mAb 52SR4 in scFv format) antigen-binding fragment (targeting tumor antigens) adapter molecule (56).
On the basis of the adapter switches, logic gates can also be introduced. For instance, the “AND” gate ensures that CAR-T cells are exclusively activated when both externally administered adapters are present, and simultaneous recognition of the two target antigens occurs (40). In the case of the A AND (NOT B) gate, the affinity of adapter A for adapter B surpasses its affinity for the receptor on T cells. Consequently, when both antigens A and B exist on normal cells, adapter B binds to adapter A, thereby impeding its ability to activate T cells (40). Cho et al. (57) developed a SUPRA CAR system capable of executing a three-input (A AND B) AND NOT C logic gate, which demonstrated exceptional controllability by conducting intricate biocomputation. The latching orthogonal cage–key protein (LOCKR) switches use the binding between the “cage protein” and the “key protein” to adopt an active conformation. This conformation, in turn, binds with CAR, allowing for the implementation of three-input (A AND B) AND NOT C logic (58). However, notably, a system constructed of exogenous substances and multitarget antigens may cause immunogenicity and a greater risk of off-target toxicity.
Small-molecule switches
By harnessing the advantages of genetic engineering and orthogonal chemical tools, researchers have developed small molecule–based safety switches. These safety switches are designed using small-molecule drugs, including proteolytic targeted chimeric (PROTAC) compounds (59), rapamycin (60–62), lenalidomide (63), dasatinib (64, 65), and grazoprevir (55). These switches can be divided into three categories: on, off, and “on/off” dual switches. Small-molecule switches not only allow flexible control over the activation and inactivation states of CAR-T cells but also have the potential to facilitate rest periods and prevent or reverse T cell exhaustion induced by tonic CAR signaling (55, 66). Moreover, through the control of drug dosage, targeting tumor cells characterized by high antigen expression while preserving healthy tissues with lower antigen expression becomes possible (55). Through comprehensive exploration of small-molecule switches, leakage activity tends to decrease when the system is turned off, accompanied by an expanding dynamic range between the on and off states.
The classical on switch system is the chemically induced dimerization (CID) system. CAR is inactive because of the dispersion between subunits, and units can dimerize to form a complete CAR. When both the recognition of the target antigen and the presence of dimeric drugs are satisfied, CAR-T cells can achieve killing. Other systems include the rapamycin-FKPB12-FRB system (60, 61), the Al120-RS3-hRBP4 system (67), and the lenalidomide-CRBN-IKZF3 system (63). Notably, rapamycin has potent immunosuppressive properties (68, 69), which may partially diminish the effectiveness of CAR-T cell therapy. Currently, there are several structurally modified rapamycin compounds aimed at reducing immunosuppressive activity (70). However, as an mTORC1 inhibitor, rapamycin has been reported to promote the accumulation of CXCR4 and potentially promote the infiltration of CAR-T cells (71). GoCAR-T cells, which target prostate stem cell antigen (PSCA) and are armed with a rimiducid-inducible MyD88/CD40 costimulation switch, demonstrated rimiducid-dependent activity (72). However, clinical trials were halted because of severe adverse reactions. Furthermore, the tet-tetracycline system serves as an on switch. After addition, tetracycline binds to the DNA binding domain rtetR, causing rtetR to recognize its binding sequence tetO and leading to activation of the promoter for the initiation of CAR expression (73, 74). In addition, Yamada et al. (75) achieved dual control of blue light and tetracycline through the Cry2-CIB1 light-induced binding switch. Sahillioglu et al. (76) developed a universal on switch for antigen receptors that has immunoreceptor tyrosine-based activation motif (ITAMs) without the need for covalent modification.
Off switches often use small molecules to degrade and inactivate the CAR. Richman et al. (77) fused CAR with the ligand-induced degradation (LID) domain, which contains a hidden degron that is exposed when specific small-molecule ligands are administered, resulting in proteasomal degradation of CAR-LID fusion proteins and inhibition of CAR-T cell activity. Juillerat et al. developed a framework controlled by proteases and protease inhibitors in which the protease target site is cleaved and CAR is preserved and functional in the absence of the protease inhibitor asunaprevir (ASN). In contrast, treatment with ASN leads to CAR degradation via a degron-mediated proteolysis pathway due to the inhibition of hepatitis C virus (HCV) nonstructural protein 3 (NS3) protease cleavage activity (78). Similarly, lenalidomide (63) or PROTAC (59) can be used to induce CAR protein hydrolysis. Moreover, other off switches, such as the tyrosine kinase inhibitor dasatinib, can reduce the cytotoxicity and proliferation capacity of CAR-T cells (64, 65). Because of its short half-life, the transition between the on and off states can be more controllable (79). In addition, Mitchell’s team found that the in situ coupling of polyethylene glycol (PEG) to the surface of CAR-T cells (via PEGylation) resulted in the formation of a polymeric spacer, which inhibited cell-to-cell interactions. This process mitigated CRS and neurotoxicity (80).
In addition, dual on/off switches can be constructed. Duong et al. (81) designed the dual-switch system using a rimiducid-induced activation switch and a rapamycin-induced apoptosis switch. This drug-responsive dual-switch system led to more controlled regulation compared to that of the single-switch system, as the dual-switch system enables the simultaneous maintenance of the “activated” state of CAR-T cells, facilitating robust proliferation and prolonged presence while also enabling the “inactivated” state, which effectively attenuates adverse effects through the integration of safety switches. However, a clinical trial of BPX-603, which was equipped with a rimiducid-inducible MyD88 and CD40 (iMC) on switch and an iCas9 suicide switch, was suspended because of toxicity issues (Table 3). In addition, Khalil and colleagues (82) developed a toolkit containing 11 programmable synthetic transcription factors. These transcription factors can be selectively activated on demand using US Food and Drug Administration (FDA)–approved small-molecule inducers to trigger specific cellular programs such as proliferation and antitumor activity. This selective activation allows for precise temporal control of CAR-T cell behavior.
Chassis cell modification
Chassis cells are the “hardware” and cornerstone of synthetic biology. Engineering the metabolic network of chassis cells allows them to achieve the desired functions. Wiebking et al. knocked out the uridine monophosphate synthase (UMPS) gene in T cells, rendering their proliferation dependent on exogenous uridine. By modulating the availability of uridine, the proliferation of T cells was effectively regulated, as evidenced by experimental findings both in vitro and in vivo (83). Furthermore, future work will involve the introduction of synthetic biology gene networks to construct chassis cells with multi-stability, which will allow CAR-T cells to mutually switch and self-regulate between various stable states (84).
THE SECURITY GUARANTEES OF CAR-T CELLS IN THE SPATIAL DIMENSION
Boolean logic gates constructed by synthetic receptors
The function of synthetic receptors depends on the outer membrane domain (determining the target cell type) and the intracellular domain (determining the signal type, positive or negative, and signal strength). Combining synthetic receptors with different functions can create Boolean logic gates such as the AND gate and “NAND” gate, thereby enabling precise tumor tissue targeting and reducing OTOT toxicity (Fig. 2).
The AND gate serves as an activation mechanism only when two distinct target antigens are recognized simultaneously. The classic AND gate is the synNotch receptor system. Unlike CAR receptors, which directly initiate T cell activation upon antigen binding, synNotch receptors recognize target antigens via extracellular recognition domains and undergo cleavage and the subsequent release of transcriptional activation domains to induce CAR expression, thereby targeting the second antigen. This system can selectively remove target cells containing both antigens to mitigate the risk of OTOT toxicity (85). Furthermore, research has shown that synNotch CAR circuits can inhibit CAR-mediated tonic signaling, thereby promoting the maintenance of long-lived memory and a nonexhausted phenotype (86). Zhu et al. developed a class of synthetic intramembrane proteolysis receptors that can optimize the sensitivity and strength of CAR-T cells, which provided modular design strategies for synNotch design. This receptor has the advantages of a compact structure, high tunability, and a low risk of immunogenicity due to the use of human proteins (87). Furthermore, the synNotch receptor can also drive the production of IL-2, thereby minimizing systemic IL-2 toxicity as much as possible (88). The potential risk of OTOT toxicity persists when healthy tissues expressing the antigens targeted by CAR are located in close proximity to tumor cells (54). Constructing synNotch CARs with high and low affinity that target the same antigen can enhance the discrimination between normal and tumor cells based on differences in antigen expression levels (89). Furthermore, a SPLIT CAR can be generated by combining a low-affinity CAR with a chimeric costimulatory receptor (CCR). In this construct, the CAR includes only the CD3ζ chain, while the CCR exclusively contains the costimulatory domain. Both receptors necessitate the simultaneous recognition of the target antigen to elicit a robust T cell response at full intensity (90). AND gates for multiple input signals have also been developed. Sukumaran et al. (91) identified three chimeric receptors: PSCA, IL-4, and transforming growth factor–β (TGF-β). Drawing on a comprehensive understanding of the complex T cell signaling network, Tousley et al. devised a Boolean logic AND-gated CAR-T cell system by combining LAT and SLP-76. This groundbreaking design represents an inaugural approach that is capable of promptly, directly, and reversibly modulating activity, and this approach is strictly confined by the recognition of dual antigens (92).
The NAND gate, also known as the A AND (NOT B) gate, is designed to activate when it identifies target cells expressing the A antigen but not the B antigen. This gate is composed of a CAR and an inhibitory CAR (iCAR) that incorporates CTLA-4 or PD-1. CAR-triggered T cell responses can be effectively inhibited after the iCAR recognizes the target antigen, and this inhibition is dynamically reversible. When the iCAR dissociates from its ligand, the activation signal of the CAR is restored, causing the T cell response to restart (93). For example, CAR targeting carcinoembryonic antigen (CEA) (94) or mesothelin (MSLN) (95) and iCAR targeting human leukocyte antigen–A2 (HLA-A2) are used to target tumor cells with down-regulated major histocompatibility complex (MHC) expression and the loss of heterozygosity (LOH) (tumor cells without A02). Witte and colleagues (96) engineered a dual-inhibitory domain CAR (DiCAR) by incorporating two immune cell inhibitory signaling domains. The purpose of this design was to finely regulate the cytotoxicity of CAR-T cells, thereby enhancing the inhibitory efficacy of CAR-T cells in comparison to that of an iCAR equipped with a single PD-1 domain. Notably, the affinity of iCAR needs to be within an appropriate range. If the affinity of the iCAR is too high and if there is a small amount of expression in tumor cells, there is a possibility of inadvertent inhibition of tumor cells as well.
CAR-T cells composed of multiple synthetic receptor Boolean logic gates can more accurately localize to tumor tissues and reduce the effect of OTOT toxicity. However, this approach also faces several challenges. First, these systems require the expression of various genes, and larger genome sizes can lead to a notable reduction in the efficiency of transgenic integration (97). Second, the potential immunogenicity of non–human-derived components, such as viral transcription factors, is a major clinical hurdle. Increasing the number of recognized antigens increases the probability of immune escape, and tumors need to change only one of the expression patterns of multiple antigens to prevent killing. Attaining an efficacious immune response against tumors entails a delicate equilibrium between safety and efficacy, necessitating the careful consideration of both factors when addressing safety concerns.
Redirected migration of T cells
In addition to the aforementioned approaches, synthetic biology techniques can also be used to genetically engineer CAR-T cells, thereby allowing for their redirected migration toward tumor sites (Fig. 2). Peng et al. (98) introduced the natural chemokine receptor C-X-C motif chemokine receptor 2 (CXCR2) into engineered T cells to make them tend to melanoma cells that expressed chemokines C-X-C motif chemokine ligand 1 (CXCL1) and C-X-C motif chemokine ligand 8 (CXCL8). Moon et al. (99) introduced the chemokine receptor C-C chemokine receptor type 2b (CCR2b) into CAR-T cells to make them tend to malignant pleural mesotheliomas (MPMs) that express the chemokine C-C motif ligand 2 (CCL2). Native receptors for natural ligands may have the problem of insufficient tumor cell specificity, which can be solved by orthogonal receptor-drug pairs composed of synthetic tropism receptors and small-molecule ligands. The synthetic tropism receptors developed by Park et al. (100) tend to bind to the bioinert small-molecule clozapine-N-oxide. Xu et al. (101) developed a photoactivatable-chemokine receptor (PA-CXCR4) to mediate light-induced chemokine signaling activation and guide cell migration to light-stimulated sites. Furthermore, Vincent et al. (102) used tumor-colonizing multifunctional bacteria (probiotics) to generate synthetic antigens within the tumor and release chemotactic factors. This approach directs CAR-T cells specifically toward the tumor site.
Targeting the tumor microenvironment
As the tumor microenvironment is characterized by hypoxia, modifying CAR-T cells to function only in a hypoxic microenvironment can substantially minimize the potential for OTOT toxicity and broaden the range of target antigens exploitable for therapeutic applications (Fig. 2). CAR-T cells can sense the hypoxic environment through hypoxia-inducible factor 1α (HIF1α) and subsequently induce CAR expression to recognize the antigen. CAR expression stops rapidly when the hypoxic environment signal is removed (103). In addition, HypoxiCAR is a dual oxygen sensing system based on the HIF1α-mediated CAR expression system in which the oxygen-dependent degradation domain (ODD) of the HIF1α is added to the CAR to achieve CAR degradation under normoxia and CAR expression under hypoxia (104). However, certain organs in the body may share similar hypoxic characteristics with the tumor microenvironment, such as the intestine (105) and kidneys (106), making it particularly important to consider these tissues when selecting targets. In addition, tumors frequently secrete proteases to facilitate invasion and promote tumor development. Incorporating a linker sensitive to tumor-specific proteases, along with a masking peptide, can hold the CAR in an inactive state. CAR-T cells are activated only when they are within the tumor microenvironment containing these specific proteases, leading to the destruction of tumor cells (107). Moreover, the products CCT301-38 and CCT301-59 transform the growth-inhibitory acidic tumor microenvironment into an activating signal, thereby reducing the risk of OTOT toxicity. These interventions are currently being investigated in the clinical trial NCT03393936 (Table 3).
THE DUAL SECURITY GUARANTEES OF CAR-T CELLS IN THE TEMPORAL AND SPATIAL DIMENSIONS
There are also methods for guaranteeing dual security in temporal and spatial dimensions, including light-operated switches and ultrasonic-operated switches (Fig. 2). Light-responsive switches offer precise control over the activation, localization, and inactivation of CAR-T cells through the regulation of the presence, distribution, and intensity of light. Zhang et al. developed an ultraviolet-sensitive photolyzable molecule (FITC-O-folate) adapter system to realize dual control by an adapter molecule and light, in which the FITC and folic acid groups are connected by a photocleavable o-nitrobenzyl ester structure. The FITC structure can bind to T cells loaded with anti-FITC receptors, and the folate structure can bind to the folate receptors of tumor cells. Exposure to 365-nm light results in the termination of CAR-T cell activation due to disruption of the photocleavable o-nitrophenyl ester structure (108). Moreover, a cleavable off switch adapter can be used to facilitate the cleavage of biotin upon exposure to ultraviolet light, thereby impeding its recognition by CAR-T cells (109). Huang et al. (110) developed a light-inducible nuclear translocation and dimerization system to achieve spatiotemporal control of CAR-T cells. Furthermore, the heat generated from light can also drive CAR expression through the utilization of a photothermal switch (111). One of the greatest challenges in using light-operated switches is the poor penetration ability of light, which may limit their practical application in deep-seated tumors or tissues. Furthermore, the restricted functionality of light-operated switches to primary or local sites may limit their application, especially considering that most patients undergoing CAR-T cell therapy typically present with metastatic diseases involving multiple and hidden metastatic sites.
Ultrasound-operated switches have similar advantages as light-operated switches, but they have the added benefit of deeper penetration. Ultrasound waves have a greater ability to penetrate tissues and reach deeper parts of the body, which limits light wave penetration. Wu et al. activated the expression of heat shock proteins through heat energy conversion by ultrasound, thereby activating the expression of CAR through the heat shock protein promoter. These switches can be directly controlled by magnetic resonance imaging (MRI)–guided focused ultrasound (FUS) without any exogenous cofactors. Short-pulse FUS stimulation enables precise and spatiotemporal control of CAR-T cells at specific locations and desired time points (112).
CONCLUSION AND FUTURE PROSPECTS
The rapid advancement of synthetic biology has facilitated the development of numerous sensors and controllable gene circuits for engineering and enhancing immune cell functions beyond the capabilities of the natural immune system. By manipulating molecular components, synthetic biology enables the robust and modular control of the cellular behavior of CAR-T cells, thereby offering innovative solutions for achieving specific immune cell functions inspired by natural systems. Overall, these functions can be regulated in a self-controlled and user-controlled paradigm, which is realized by automatic cell execution behavior (such as automatic activation upon recognition of target cells) and human manipulation through external factors (such as the utilization of small molecules and light). On the basis of engineering, modular, and systematic characteristics, synthetic biology is undoubtedly of great value in enhancing safety. Recent efforts have concentrated on applying synthetic biology methods to address safety concerns in CAR-T cell therapy across the temporal and spatial dimensions (Fig. 3). Numerous synthetic biology approaches involving CAR-T cells have demonstrated promising outcomes in clinical trials. As synthetic biology continues to develop and technology matures, we anticipate broader clinical applications in the coming years.
Fig. 3. The toxicity of CAR-T cell therapy hinders its further application.
In this review, we categorize the adverse effects associated with CAR-T cell therapy into two domains and provide a comprehensive overview of recent advancements in using synthetic biology approaches to address safety concerns. Figure created with BioRender.com.
Several challenges remain in the application of synthetic biology to enhance the safety of CAR-T cells. First, the immunogenicity caused by nonhuman substances, such as nonhuman transcription factors or proteins with specific functions derived from viruses, bacteria, or other animals and plants, is still worthy of vigilance. Vigilance is needed to explore and construct human-derived synthetic systems that minimize the risk of immune rejection. Second, there is a risk of expression leakage in modified CAR-T cell gene circuits, which is a common concern in synthetic biology–based gene expression regulation models. In CAR-T cell design, gene circuits are constructed by applying different biological elements, and design-build-test-learning is performed to reduce the leakage of expressed genes. In addition, ensuring that the introduced gene circuit is biologically orthogonal to the endogenous T cell response is crucial. Last, the delivery and integration of complex gene circuits in synthetic biology modifications face technical limitations, thereby imposing increased burdens and technical requirements on industrial production. Currently, gene delivery systems for CAR-T cell production use both viral and nonviral delivery systems. Viral delivery systems have limitations in terms of their gene payload and pose a risk of insertional mutagenesis. Nonviral delivery systems, such as piggyBac, Sleeping Beauty, and the Tol2 transposon system, offer lower production costs, a reduced risk of insertional mutagenesis, and larger gene loads. However, these methods demonstrate lower gene transfer efficiency and require longer in vitro cultivation. Future efforts should concentrate on exploring new methods to increase gene delivery/transduction efficiency and load to facilitate the industrial production of CAR-T cells equipped with synthetic biology switches.
In the future, CAR-T cells are expected to advance toward more sophisticated engineered cells, a goal that can be achieved with the wide variety of components, logic gates, and genetic circuits provided by synthetic biology approaches. In addition, the development of universal CAR-T cell therapy is a future research focus. Compared with conventional CAR-T cells, universal CAR-T cells may cause more serious adverse reactions, such as GVHD and immunological rejection. Therefore, the forthcoming stages of research will prioritize the modification of universal chassis cells to enhance safety. Moreover, thorough investigations of the adverse effects of CAR-T cell therapy are urgently needed. Clinical trials of BPX-601 and BPX-603, which included additional switches, were stopped because of severe adverse effects. The FDA has also announced additional inquiries into the risk of secondary T cell malignancies linked to CAR-T cell therapy (113). Studies further indicate that CAR-T cell therapy may activate dormant viruses, which can lead to severe complications (114). These findings underscore the persistent need for a comprehensive assessment of safety concerns related to CAR-T cell therapy. In summary, as a highly promising antitumor cell therapy, CAR-T cell therapy exhibits remarkable potential for treating solid tumors and hematological malignancies. We anticipate the emergence of safer, more controllable, more intelligent, and universal CAR-T cell therapy in future clinical treatments.
Acknowledgments
Figures for this article are created with BioRender.com
Funding: This work was supported by grants from Zhejiang Provincial Natural Science Foundation (no. LR22H310002 to J.C.), the Fundamental Research Funds for the Central Universities (no. 226-2023-00059), the Zhejiang University K.P.Chao’s High Technology Development Foundation, and the National Natural Science Foundation of China (82204413).
Author contributions: L.L.: Writing—original draft, conceptualization, writing—review and editing, funding acquisition, supervision, project administration, and visualization. M.X.: Writing—original draft, conceptualization, and validation. B.Y.: Writing—original draft, conceptualization, writing—review and editing, funding acquisition, supervision, project administration, and visualization. W.-b.Z.: Writing—original draft, conceptualization, writing—review and editing, funding acquisition, supervision, project administration, and visualization. J.C.: Writing—original draft, conceptualization, writing—review and editing, funding acquisition, supervision, project administration, and visualization.
Competing interests: The authors declare that they have no competing interests.
Data and materials availability: All data needed to evaluate the conclusions in the paper are present in the paper.
REFERENCES AND NOTES
- 1.Yang F., Jin H., Wang J., Sun Q., Yan C., Wei F., Ren X., Adoptive cellular therapy (ACT) for cancer treatment. Adv. Exp. Med. Biol. 909, 169–239 (2016). [DOI] [PubMed] [Google Scholar]
- 2.Le R. Q., Li L., Yuan W., Shord S. S., Nie L., Habtemariam B. A., Przepiorka D., Farrell A. T., Pazdur R., FDA approval summary: Tocilizumab for treatment of chimeric antigen receptor T cell-induced severe or life-threatening cytokine release syndrome. Oncologist 23, 943–947 (2018). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 3.Park J. H., Nath K., Devlin S. M., Sauter C. S., Palomba M. L., Shah G., Dahi P., Lin R. J., Scordo M., Perales M. A., Shouval R., Tomas A. A., Cathcart E., Mead E., Santomasso B., Holodny A., Brentjens R. J., Riviere I., Sadelain M., CD19 CAR T-cell therapy and prophylactic anakinra in relapsed or refractory lymphoma: Phase 2 trial interim results. Nat. Med. 29, 1710–1717 (2023). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 4.Wu C. Y., Rupp L. J., Roybal K. T., Lim W. A., Synthetic biology approaches to engineer T cells. Curr. Opin. Immunol. 35, 123–130 (2015). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 5.Frey N., Porter D., Cytokine release syndrome with chimeric antigen receptor T cell therapy. Biol. Blood Marrow Transplant. 25, e123–e127 (2019). [DOI] [PubMed] [Google Scholar]
- 6.Mirzaee Godarzee M., Mahmud Hussen B., Razmara E., Hakak-Zargar B., Mohajerani F., Dabiri H., Fatih Rasul M., Ghazimoradi M. H., Babashah S., Sadeghizadeh M., Strategies to overcome the side effects of chimeric antigen receptor T cell therapy. Ann. N. Y. Acad. Sci. 1510, 18–35 (2022). [DOI] [PubMed] [Google Scholar]
- 7.Lee D. W., Gardner R., Porter D. L., Louis C. U., Ahmed N., Jensen M., Grupp S. A., Mackall C. L., Current concepts in the diagnosis and management of cytokine release syndrome. Blood 124, 188–195 (2014). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 8.Shimabukuro-Vornhagen A., Godel P., Subklewe M., Stemmler H. J., Schlosser H. A., Schlaak M., Kochanek M., Boll B., von Bergwelt-Baildon M. S., Cytokine release syndrome. J. Immunother. Cancer 6, 56 (2018). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 9.Santomasso B. D., Park J. H., Salloum D., Riviere I., Flynn J., Mead E., Halton E., Wang X., Senechal B., Purdon T., Cross J. R., Liu H., Vachha B., Chen X., DeAngelis L. M., Li D., Bernal Y., Gonen M., Wendel H. G., Sadelain M., Brentjens R. J., Clinical and biological correlates of neurotoxicity associated with CAR T-cell therapy in patients with B-cell acute lymphoblastic leukemia. Cancer Discov. 8, 958–971 (2018). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 10.Sterner R. C., Sterner R. M., Immune effector cell associated neurotoxicity syndrome in chimeric antigen receptor-T cell therapy. Front. Immunol. 13, 879608 (2022). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 11.Harrison R. A., Majd N. K., Tummala S., de Groot J. F., Neurologic toxicities of immunotherapy. Adv. Exp. Med. Biol. 1342, 417–429 (2021). [DOI] [PubMed] [Google Scholar]
- 12.Wang Z., Han W., Biomarkers of cytokine release syndrome and neurotoxicity related to CAR-T cell therapy. Biomark. Res. 6, 4 (2018). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 13.Teachey D. T., Lacey S. F., Shaw P. A., Melenhorst J. J., Maude S. L., Frey N., Pequignot E., Gonzalez V. E., Chen F., Finklestein J., Barrett D. M., Weiss S. L., Fitzgerald J. C., Berg R. A., Aplenc R., Callahan C., Rheingold S. R., Zheng Z., Rose-John S., White J. C., Nazimuddin F., Wertheim G., Levine B. L., June C. H., Porter D. L., Grupp S. A., Identification of predictive biomarkers for cytokine release syndrome after chimeric antigen receptor T-cell therapy for acute lymphoblastic leukemia. Cancer Discov. 6, 664–679 (2016). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 14.Butt O. H., Zhou A. Y., Ances B. M., DiPersio J. F., Ghobadi A., A systematic framework for predictive biomarkers in immune effector cell-associated neurotoxicity syndrome. Front. Neurol. 14, 1110647 (2023). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 15.Schumacher T. N., Schreiber R. D., Neoantigens in cancer immunotherapy. Science 348, 69–74 (2015). [DOI] [PubMed] [Google Scholar]
- 16.Uhlen M., Fagerberg L., Hallstrom B. M., Lindskog C., Oksvold P., Mardinoglu A., Sivertsson A., Kampf C., Sjostedt E., Asplund A., Olsson I., Edlund K., Lundberg E., Navani S., Szigyarto C. A., Odeberg J., Djureinovic D., Takanen J. O., Hober S., Alm T., Edqvist P. H., Berling H., Tegel H., Mulder J., Rockberg J., Nilsson P., Schwenk J. M., Hamsten M., von Feilitzen K., Forsberg M., Persson L., Johansson F., Zwahlen M., von Heijne G., Nielsen J., Ponten F., Tissue-based map of the human proteome. Science 347, 1260419 (2015). [DOI] [PubMed] [Google Scholar]
- 17.Uhlen M., Oksvold P., Fagerberg L., Lundberg E., Jonasson K., Forsberg M., Zwahlen M., Kampf C., Wester K., Hober S., Wernerus H., Bjorling L., Ponten F., Towards a knowledge-based human protein atlas. Nat. Biotechnol. 28, 1248–1250 (2010). [DOI] [PubMed] [Google Scholar]
- 18.Lamers C. H., Sleijfer S., Vulto A. G., Kruit W. H., Kliffen M., Debets R., Gratama J. W., Stoter G., Oosterwijk E., Treatment of metastatic renal cell carcinoma with autologous T-lymphocytes genetically retargeted against carbonic anhydrase IX: First clinical experience. J. Clin. Oncol. 24, e20–e22 (2006). [DOI] [PubMed] [Google Scholar]
- 19.Lamers C. H., Klaver Y., Gratama J. W., Sleijfer S., Debets R., Treatment of metastatic renal cell carcinoma (mRCC) with CAIX CAR-engineered T-cells-a completed study overview. Biochem. Soc. Trans. 44, 951–959 (2016). [DOI] [PubMed] [Google Scholar]
- 20.Feng K., Liu Y., Guo Y., Qiu J., Wu Z., Dai H., Yang Q., Wang Y., Han W., Phase I study of chimeric antigen receptor modified T cells in treating HER2-positive advanced biliary tract cancers and pancreatic cancers. Protein Cell 9, 838–847 (2018). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 21.Feng K. C., Guo Y. L., Liu Y., Dai H. R., Wang Y., Lv H. Y., Huang J. H., Yang Q. M., Han W. D., Cocktail treatment with EGFR-specific and CD133-specific chimeric antigen receptor-modified T cells in a patient with advanced cholangiocarcinoma. J. Hematol. Oncol. 10, 4 (2017). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 22.Guo Y., Feng K., Liu Y., Wu Z., Dai H., Yang Q., Wang Y., Jia H., Han W., Phase I study of chimeric antigen receptor-modified T cells in patients with EGFR-positive advanced biliary tract cancers. Clin. Cancer Res. 24, 1277–1286 (2018). [DOI] [PubMed] [Google Scholar]
- 23.Liu Y., Guo Y., Wu Z., Feng K., Tong C., Wang Y., Dai H., Shi F., Yang Q., Han W., Anti-EGFR chimeric antigen receptor-modified T cells in metastatic pancreatic carcinoma: A phase I clinical trial. Cytotherapy 22, 573–580 (2020). [DOI] [PubMed] [Google Scholar]
- 24.Zhou X., Dotti G., Krance R. A., Martinez C. A., Naik S., Kamble R. T., Durett A. G., Dakhova O., Savoldo B., Di Stasi A., Spencer D. M., Lin Y. F., Liu H., Grilley B. J., Gee A. P., Rooney C. M., Heslop H. E., Brenner M. K., Inducible caspase-9 suicide gene controls adverse effects from alloreplete T cells after haploidentical stem cell transplantation. Blood 125, 4103–4113 (2015). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 25.Di Stasi A., Tey S. K., Dotti G., Fujita Y., Kennedy-Nasser A., Martinez C., Straathof K., Liu E., Durett A. G., Grilley B., Liu H., Cruz C. R., Savoldo B., Gee A. P., Schindler J., Krance R. A., Heslop H. E., Spencer D. M., Rooney C. M., Brenner M. K., Inducible apoptosis as a safety switch for adoptive cell therapy. N. Engl. J. Med. 365, 1673–1683 (2011). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 26.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., Li Pira G., Leone G., Bertaina V., Sinibaldi M., Di Cecca S., Guercio M., Abbaszadeh Z., Iaffaldano L., Gunetti M., Iacovelli S., Bugianesi R., Macchia S., Algeri M., Merli P., Galaverna F., Abbas R., Garganese M. C., Villani M. F., Colafati G. S., Bonetti F., Rabusin M., Perruccio K., Folsi V., Quintarelli C., Locatelli F., I. O. P. B. G., GD2-CART01 for relapsed or refractory high-risk neuroblastoma. N. Engl. J. Med. 388, 1284–1295 (2023). [DOI] [PubMed] [Google Scholar]
- 27.Sasaki T., Sakoda Y., Adachi K., Tokunaga Y., Tamada K., Therapeutic effects of anti-GM2 CAR-T cells expressing IL-7 and CCL19 for GM2-positive solid cancer in xenograft model. Cancer Med. 12, 12569–12580 (2023). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 28.Berger C., Flowers M. E., Warren E. H., Riddell S. R., Analysis of transgene-specific immune responses that limit the in vivo persistence of adoptively transferred HSV-TK-modified donor T cells after allogeneic hematopoietic cell transplantation. Blood 107, 2294–2302 (2006). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 29.Marin V., Cribioli E., Philip B., Tettamanti S., Pizzitola I., Biondi A., Biagi E., Pule M., Comparison of different suicide-gene strategies for the safety improvement of genetically manipulated T cells. Hum. Gene Ther. Methods 23, 376–386 (2012). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 30.Haberkorn U., Altmann A., Morr I., Germann C., Oberdorfer F., van Kaick G., Multitracer studies during gene therapy of hepatoma cells with herpes simplex virus thymidine kinase and ganciclovir. J. Nucl. Med. 38, 1048–1054 (1997). [PubMed] [Google Scholar]
- 31.Keu K. V., Witney T. H., Yaghoubi S., Rosenberg J., Kurien A., Magnusson R., Williams J., Habte F., Wagner J. R., Forman S., Brown C., Allen-Auerbach M., Czernin J., Tang W., Jensen M. C., Badie B., Gambhir S. S., Reporter gene imaging of targeted T cell immunotherapy in recurrent glioma. Sci. Transl. Med. 9, eaag2196 (2017). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 32.Shin J., Parker M. F. L., Zhu I., Alanizi A., Rodriguez C. I., Liu R., Watchmaker P. B., Kalita M., Blecha J., Luu J., Wright B., Lapi S. E., Flavell R. R., Okada H., Tlsty T. D., Roybal K. T., Wilson D. M., Antigen-dependent inducible T-cell reporter system for PET imaging of breast cancer and glioblastoma. J. Nucl. Med. 64, 137–144 (2023). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 33.Abramson J. S., Palomba L., Gordon L. I., Lunning M., Arnason J., Forero-Torres A., Albertson T. M., Exton V. S., Sutherland C., Xie B., Snodgrass S., Siddiqi T., Transcend NHL 001: Immunotherapy with the CD19-directed CAR T-cell product JCAR017 results in high complete response rates in relapsed or refractory B-cell non-Hodgkin lymphoma. Blood 128, 4192 (2016). [Google Scholar]
- 34.Tran V. L., Novell A., Tournier N., Gerstenmayer M., Schweitzer-Chaput A., Mateos C., Jego B., Bouleau A., Nozach H., Winkeler A., Kuhnast B., Larrat B., Truillet C., Impact of blood-brain barrier permeabilization induced by ultrasound associated to microbubbles on the brain delivery and kinetics of cetuximab: An immunoPET study using 89Zr-cetuximab. J. Control. Release 328, 304–312 (2020). [DOI] [PubMed] [Google Scholar]
- 35.Amini L., Silbert S. K., Maude S. L., Nastoupil L. J., Ramos C. A., Brentjens R. J., Sauter C. S., Shah N. N., Abou-El-Enein M., Preparing for CAR T cell therapy: Patient selection, bridging therapies and lymphodepletion. Nat. Rev. Clin. Oncol. 19, 342–355 (2022). [DOI] [PubMed] [Google Scholar]
- 36.Bouquet L., Bole-Richard E., Warda W., Neto Da Rocha M., Trad R., Nicod C., Haderbache R., Genin D., Ferrand C., Deschamps M., RapaCaspase-9-based suicide gene applied to the safety of IL-1RAP CAR-T cells. Gene Ther. 30, 706–713 (2023). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 37.Gorovits B., Koren E., Immunogenicity of chimeric antigen receptor T-cell therapeutics. BioDrugs 33, 275–284 (2019). [DOI] [PubMed] [Google Scholar]
- 38.Zhou X., Di Stasi A., Tey S. K., Krance R. A., Martinez C., Leung K. S., Durett A. G., Wu M. F., Liu H., Leen A. M., Savoldo B., Lin Y. F., Grilley B. J., Gee A. P., Spencer D. M., Rooney C. M., Heslop H. E., Brenner M. K., Dotti G., Long-term outcome after haploidentical stem cell transplant and infusion of T cells expressing the inducible caspase 9 safety transgene. Blood 123, 3895–3905 (2014). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 39.Lee Y. G., Chu H., Lu Y., Leamon C. P., Srinivasarao M., Putt K. S., Low P. S., Regulation of CAR T cell-mediated cytokine release syndrome-like toxicity using low molecular weight adapters. Nat. Commun. 10, 2681 (2019). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 40.Cho J. H., Collins J. J., Wong W. W., Universal chimeric antigen receptors for multiplexed and logical control of T cell responses. Cell 173, 1426–1438.e11 (2018). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 41.Chen Y. Y., Increasing T cell wersatility with SUPRA CARs. Cell 173, 1316–1317 (2018). [DOI] [PubMed] [Google Scholar]
- 42.Tamada K., Geng D., Sakoda Y., Bansal N., Srivastava R., Li Z., Davila E., Redirecting gene-modified T cells toward various cancer types using tagged antibodies. Clin. Cancer Res. 18, 6436–6445 (2012). [DOI] [PubMed] [Google Scholar]
- 43.Lee Y. G., Marks I., Srinivasarao M., Kanduluru A. K., Mahalingam S. M., Liu X., Chu H., Low P. S., Use of a single CAR T cell and several bispecific adapters racilitates eradication of multiple antigenically different solid tumors. Cancer Res. 79, 387–396 (2019). [DOI] [PubMed] [Google Scholar]
- 44.Zhang B., Wang Y., Yuan Y., Sun J., Liu L., Huang D., Hu J., Wang M., Li S., Song W., Chen H., Zhou D., Zhang X., In vitro elimination of autoreactive B cells from rheumatoid arthritis patients by universal chimeric antigen receptor T cells. Ann. Rheum. Dis. 80, 176–184 (2021). [DOI] [PubMed] [Google Scholar]
- 45.Kim M. S., Ma J. S., Yun H., Cao Y., Kim J. Y., Chi V., Wang D., Woods A., Sherwood L., Caballero D., Gonzalez J., Schultz P. G., Young T. S., Kim C. H., Redirection of genetically engineered CAR-T cells using bifunctional small molecules. J. Am. Chem. Soc. 137, 2832–2835 (2015). [DOI] [PubMed] [Google Scholar]
- 46.Cao Y., Rodgers D. T., Du J., Ahmad I., Hampton E. N., Ma J. S., Mazagova M., Choi S. H., Yun H. Y., Xiao H., Yang P., Luo X., Lim R. K., Pugh H. M., Wang F., Kazane S. A., Wright T. M., Kim C. H., Schultz P. G., Young T. S., Design of switchable chimeric antigen receptor T cells targeting breast cancer. Angew. Chem. Int. Ed. Engl. 55, 7520–7524 (2016). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 47.Urbanska K., Lanitis E., Poussin M., Lynn R. C., Gavin B. P., Kelderman S., Yu J., Scholler N., Powell D. J. Jr., A universal strategy for adoptive immunotherapy of cancer through use of a novel T-cell antigen receptor. Cancer Res. 72, 1844–1852 (2012). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 48.Lohmueller J. J., Ham J. D., Kvorjak M., Finn O. J., mSA2 affinity-enhanced biotin-binding CAR T cells for universal tumor targeting. Oncoimmunology 7, e1368604 (2017). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 49.Viaud S., Ma J. S. Y., Hardy I. R., Hampton E. N., Benish B., Sherwood L., Nunez V., Ackerman C. J., Khialeeva E., Weglarz M., Lee S. C., Woods A. K., Young T. S., Switchable control over in vivo CAR T expansion, B cell depletion, and induction of memory. Proc. Natl. Acad. Sci. U.S.A. 115, E10898–E10906 (2018). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 50.Rodgers D. T., Mazagova M., Hampton E. N., Cao Y., Ramadoss N. S., Hardy I. R., Schulman A., Du J., Wang F., Singer O., Ma J., Nunez V., Shen J., Woods A. K., Wright T. M., Schultz P. G., Kim C. H., Young T. S., Switch-mediated activation and retargeting of CAR-T cells for B-cell malignancies. Proc. Natl. Acad. Sci. U.S.A. 113, E459–E468 (2016). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 51.Stepanov A. V., Kalinin R. S., Shipunova V. O., Zhang D., Xie J., Rubtsov Y. P., Ukrainskaya V. M., Schulga A., Konovalova E. V., Volkov D. V., Yaroshevich I. A., Moysenovich A. M., Belogurov A. A. Jr., Zhang H., Telegin G. B., Chernov A. S., Maschan M. A., Terekhov S. S., Wu P., Deyev S. M., Lerner R. A., Gabibov A. G., Altman S., Switchable targeting of solid tumors by BsCAR T cells. Proc. Natl. Acad. Sci. U.S.A. 119, e2210562119 (2022). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 52.Loff S., Dietrich J., Meyer J. E., Riewaldt J., Spehr J., von Bonin M., Grunder C., Swayampakula M., Franke K., Feldmann A., Bachmann M., Ehninger G., Ehninger A., Cartellieri M., Rapidly switchable universal CAR-T cells for treatment of CD123-positive leukemia. Mol. Ther. Oncolytics 17, 408–420 (2020). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 53.Meyer J. E., Loff S., Dietrich J., Spehr J., Jurado Jimenez G., von Bonin M., Ehninger G., Cartellieri M., Ehninger A., Evaluation of switch-mediated costimulation in trans on universal CAR-T cells (UniCAR) targeting CD123-positive AML. Onco. Targets Ther. 10, 1945804 (2021). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 54.Srivastava S., Salter A. I., Liggitt D., Yechan-Gunja S., Sarvothama M., Cooper K., Smythe K. S., Dudakov J. A., Pierce R. H., Rader C., Riddell S. R., Logic-gated ROR1 chimeric antigen receptor expression rescues T cell-mediated toxicity to normal tissues and enables selective tumor targeting. Cancer Cell 35, 489–503.e8 (2019). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 55.Labanieh L., Majzner R. G., Klysz D., Sotillo E., Fisher C. J., Vilches-Moure J. G., Pacheco K. Z. B., Malipatlolla M., Xu P., Hui J. H., Murty T., Theruvath J., Mehta N., Yamada-Hunter S. A., Weber E. W., Heitzeneder S., Parker K. R., Satpathy A. T., Chang H. Y., Lin M. Z., Cochran J. R., Mackall C. L., Enhanced safety and efficacy of protease-regulated CAR-T cell receptors. Cell 185, 1745–1763.e22 (2022). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 56.Peng H., Nerreter T., Mestermann K., Wachter J., Chang J., Hudecek M., Rader C., ROR1-targeting switchable CAR-T cells for cancer therapy. Oncogene 41, 4104–4114 (2022). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 57.Cho J. H., Okuma A., Sofjan K., Lee S., Collins J. J., Wong W. W., Engineering advanced logic and distributed computing in human CAR immune cells. Nat. Commun. 12, 792 (2021). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 58.Lajoie M. J., Boyken S. E., Salter A. I., Bruffey J., Rajan A., Langan R. A., Olshefsky A., Muhunthan V., Bick M. J., Gewe M., Quijano-Rubio A., Johnson J., Lenz G., Nguyen A., Pun S., Correnti C. E., Riddell S. R., Baker D., Designed protein logic to target cells with precise combinations of surface antigens. Science 369, 1637–1643 (2020). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 59.Lee S. M., Kang C. H., Choi S. U., Kim Y., Hwang J. Y., Jeong H. G., Park C. H., A chemical switch system to modulate chimeric entigen receptor T cell activity through proteolysis-targeting chimaera technology. ACS Synth. Biol. 9, 987–992 (2020). [DOI] [PubMed] [Google Scholar]
- 60.Wu C. Y., Roybal K. T., Puchner E. M., Onuffer J., Lim W. A., Remote control of therapeutic T cells through a small molecule-gated chimeric receptor. Science 350, aab4077 (2015). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 61.Leung W. H., Gay J., Martin U., Garrett T. E., Horton H. M., Certo M. T., Blazar B. R., Morgan R. A., Gregory P. D., Jarjour J., Astrakhan A., Sensitive and adaptable pharmacological control of CAR T cells through extracellular receptor dimerization. JCI Insight 5, e124430 (2019). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 62.Juillerat A., Marechal A., Filhol J. M., Valton J., Duclert A., Poirot L., Duchateau P., Design of chimeric antigen receptors with integrated controllable transient functions. Sci. Rep. 6, 18950 (2016). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 63.Jan M., Scarfo I., Larson R. C., Walker A., Schmidts A., Guirguis A. A., Gasser J. A., Slabicki M., Bouffard A. A., Castano A. P., Kann M. C., Cabral M. L., Tepper A., Grinshpun D. E., Sperling A. S., Kyung T., Sievers Q. L., Birnbaum M. E., Maus M. V., Ebert B. L., Reversible ON- and OFF-switch chimeric antigen receptors controlled by lenalidomide. Sci. Transl. Med. 13, eabb6295 (2021). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 64.Weber E. W., Lynn R. C., Sotillo E., Lattin J., Xu P., Mackall C. L., Pharmacologic control of CAR-T cell function using dasatinib. Blood Adv. 3, 711–717 (2019). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 65.Mestermann K., Giavridis T., Weber J., Rydzek J., Frenz S., Nerreter T., Mades A., Sadelain M., Einsele H., Hudecek M., The tyrosine kinase inhibitor dasatinib acts as a pharmacologic on/off switch for CAR T cells. Sci. Transl. Med. 11, eaau5907 (2019). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 66.Weber E. W., Parker K. R., Sotillo E., Lynn R. C., Anbunathan H., Lattin J., Good Z., Belk J. A., Daniel B., Klysz D., Malipatlolla M., Xu P., Bashti M., Heitzeneder S., Labanieh L., Vandris P., Majzner R. G., Qi Y., Sandor K., Chen L. C., Prabhu S., Gentles A. J., Wandless T. J., Satpathy A. T., Chang H. Y., Mackall C. L., Transient rest restores functionality in exhausted CAR-T cells through epigenetic remodeling. Science 372, eaba1786 (2021). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 67.Zajc C. U., Dobersberger M., Schaffner I., Mlynek G., Puhringer D., Salzer B., Djinovic-Carugo K., Steinberger P., De Sousa Linhares A., Yang N. J., Obinger C., Holter W., Traxlmayr M. W., Lehner M., A conformation-specific ON-switch for controlling CAR T cells with an orally available drug. Proc. Natl. Acad. Sci. U.S.A. 117, 14926–14935 (2020). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 68.Law B. K., Rapamycin: An anti-cancer immunosuppressant? Crit. Rev. Oncol. Hematol. 56, 47–60 (2005). [DOI] [PubMed] [Google Scholar]
- 69.Gollomp K., Teachey D. T., TORing the impact of sirolimus on immune health. Blood 141, 212–214 (2023). [DOI] [PubMed] [Google Scholar]
- 70.Bayle J. H., Grimley J. S., Stankunas K., Gestwicki J. E., Wandless T. J., Crabtree G. R., Rapamycin analogs with differential binding specificity permit orthogonal control of protein activity. Chem. Biol. 13, 99–107 (2006). [DOI] [PubMed] [Google Scholar]
- 71.Nian Z., Zheng X., Dou Y., Du X., Zhou L., Fu B., Sun R., Tian Z., Wei H., Rapamycin pretreatment rescues the bone marrow AML cell elimination capacity of CAR-T cells. Clin. Cancer Res. 27, 6026–6038 (2021). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 72.Stein M. N., Teply B. A., Gergis U., Strickland D., Senesac J., Bayle H., Chatwal M. S., Bilen M. A., Stadler W. M., Dumbrava E. E., Early results from a phase 1, multicenter trial of PSCA-specific GoCAR T cells (BPX-601) in patients with metastatic castration-resistant prostate cancer (mCRPC). J. Clin. Oncol. 41, 140 (2023). [Google Scholar]
- 73.Zhang R. Y., Wei D., Liu Z. K., Yong Y. L., Wei W., Zhang Z. Y., Lv J. J., Zhang Z., Chen Z. N., Bian H., Doxycycline inducible chimeric antigen receptor T cells targeting CD147 for hepatocellular carcinoma therapy. Front. Cell Dev. Biol. 7, 233 (2019). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 74.Das A. T., Tenenbaum L., Berkhout B., Tet-on systems for doxycycline-inducible gene expression. Curr. Gene Ther. 16, 156–167 (2016). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 75.Yamada M., Suzuki Y., Nagasaki S. C., Okuno H., Imayoshi I., Light control of the Tet gene expression system in mammalian cells. Cell Rep. 25, 487–500.e6 (2018). [DOI] [PubMed] [Google Scholar]
- 76.Sahillioglu A. C., Toebes M., Apriamashvili G., Gomez R., Schumacher T. N., CRASH-IT switch enables reversible and dose-dependent control of TCR and CAR T-cell function. Cancer Immunol. Res. 9, 999–1007 (2021). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 77.Richman S. A., Wang L. C., Moon E. K., Khire U. R., Albelda S. M., Milone M. C., Ligand-induced degradation of a CAR permits reversible remote control of CAR T Cell activity in vitro and in vivo. Mol. Ther. 28, 1600–1613 (2020). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 78.Juillerat A., Tkach D., Busser B. W., Temburni S., Valton J., Duclert A., Poirot L., Depil S., Duchateau P., Modulation of chimeric antigen receptor surface expression by a small molecule switch. BMC Biotechnol. 19, 44 (2019). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 79.Christopher L. J., Cui D., Wu C., Luo R., Manning J. A., Bonacorsi S. J., Lago M., Allentoff A., Lee F. Y., McCann B., Galbraith S., Reitberg D. P., He K., Barros A. Jr., Blackwood-Chirchir A., Humphreys W. G., Iyer R. A., Metabolism and disposition of dasatinib after oral administration to humans. Drug Metab. Dispos. 36, 1357–1364 (2008). [DOI] [PubMed] [Google Scholar]
- 80.Gong N., Han X., Xue L., El-Mayta R., Metzloff A. E., Billingsley M. M., Hamilton A. G., Mitchell M. J., In situ PEGylation of CAR T cells alleviates cytokine release syndrome and neurotoxicity. Nat. Mater. 22, 1571–1580 (2023). [DOI] [PubMed] [Google Scholar]
- 81.Duong M. T., Collinson-Pautz M. R., Morschl E., Lu A., Szymanski S. P., Zhang M., Brandt M. E., Chang W. C., Sharp K. L., Toler S. M., Slawin K. M., Foster A. E., Spencer D. M., Bayle J. H., Two-dimensional regulation of CAR-T cell therapy with orthogonal switches. Mol. Ther. Oncolytics 12, 124–137 (2019). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 82.Li H. S., Israni D. V., Gagnon K. A., Gan K. A., Raymond M. H., Sander J. D., Roybal K. T., Joung J. K., Wong W. W., Khalil A. S., Multidimensional control of therapeutic human cell function with synthetic gene circuits. Science 378, 1227–1234 (2022). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 83.Wiebking V., Patterson J. O., Martin R., Chanda M. K., Lee C. M., Srifa W., Bao G., Porteus M. H., Metabolic engineering generates a transgene-free safety switch for cell therapy. Nat. Biotechnol. 38, 1441–1450 (2020). [DOI] [PubMed] [Google Scholar]
- 84.Zhu R., Del Rio-Salgado J. M., Garcia-Ojalvo J., Elowitz M. B., Synthetic multistability in mammalian cells. Science 375, eabg9765 (2022). [DOI] [PubMed] [Google Scholar]
- 85.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 164, 770–779 (2016). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 86.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. 13, eabd8836 (2021). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 87.Zhu I., Liu R., Garcia J. M., Hyrenius-Wittsten A., Piraner D. I., Alavi J., Israni D. V., Liu B., Khalil A. S., Roybal K. T., Modular design of synthetic receptors for programmed gene regulation in cell therapies. Cell 185, 1431–1443.e16 (2022). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 88.Allen G. M., Frankel N. W., Reddy N. R., Bhargava H. K., Yoshida M. A., Stark S. R., Purl M., Lee J., Yee J. L., Yu W., Li A. W., Garcia K. C., El-Samad H., Roybal K. T., Spitzer M. H., Lim W. A., Synthetic cytokine circuits that drive T cells into immune-excluded tumors. Science 378, eaba1624 (2022). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 89.Hernandez-Lopez R. A., Yu W., Cabral K. A., Creasey O. A., Lopez Pazmino M. D. P., Tonai Y., De Guzman A., Makela A., Saksela K., Gartner Z. J., Lim W. A., T cell circuits that sense antigen density with an ultrasensitive threshold. Science 371, 1166–1171 (2021). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 90.Kloss C. C., Condomines M., Cartellieri M., Bachmann M., Sadelain M., Combinatorial antigen recognition with balanced signaling promotes selective tumor eradication by engineered T cells. Nat. Biotechnol. 31, 71–75 (2013). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 91.Sukumaran S., Watanabe N., Bajgain P., Raja K., Mohammed S., Fisher W. E., Brenner M. K., Leen A. M., Vera J. F., Enhancing the potency and specificity of engineered T cells for cancer treatment. Cancer Discov. 8, 972–987 (2018). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 92.Tousley A. M., Rotiroti M. C., Labanieh L., Rysavy L. W., Kim W. J., Lareau C., Sotillo E., Weber E. W., Rietberg S. P., Dalton G. N., Yin Y., Klysz D., Xu P., de la Serna E. L., Dunn A. R., Satpathy A. T., Mackall C. L., Majzner R. G., Co-opting signalling molecules enables logic-gated control of CAR T cells. Nature 615, 507–516 (2023). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 93.Fedorov V. D., Themeli M., Sadelain M., PD-1- and CTLA-4-based inhibitory chimeric antigen receptors (iCARs) divert off-target immunotherapy responses. Sci. Transl. Med. 5, 215ra172 (2013). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 94.Sandberg M. L., Wang X., Martin A. D., Nampe D. P., Gabrelow G. B., Li C. Z., McElvain M. E., Lee W. H., Shafaattalab S., Martire S., Fisher F. A., Ando Y., Liu E., Ju D., Wong L. M., Xu H., Kamb A., A carcinoembryonic antigen-specific cell therapy selectively targets tumor cells with HLA loss of heterozygosity in vitro and in vivo. Sci. Transl. Med. 14, eabm0306 (2022). [DOI] [PubMed] [Google Scholar]
- 95.Tokatlian T., Asuelime G. E., Mock J. Y., DiAndreth B., Sharma S., Warshaviak D. T., Daris M. E., Bolanos K., Luna B. L., Naradikian M. S., Deshmukh K., Hamburger A. E., Kamb A., Mesothelin-specific CAR-T cell therapy that incorporates an HLA-gated safety mechanism selectively kills tumor cells. J. Immunother. Cancer 10, e003826 (2022). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 96.Bangayan N. J., Wang L., Burton Sojo G., Noguchi M., Cheng D., Ta L., Gunn D., Mao Z., Liu S., Yin Q., Riedinger M., Li K., Wu A. M., Stoyanova T., Witte O. N., Dual-inhibitory domain iCARs improve the efficiency of the AND-NOT gate CAR T strategy. Proc. Natl. Acad. Sci. U.S.A. 120, e2312374120 (2023). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 97.Sweeney N. P., Vink C. A., The impact of lentiviral vector genome size and producer cell genomic to gag-pol mRNA ratios on packaging efficiency and titre. Mol. Ther. Methods Clin. Dev. 21, 574–584 (2021). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 98.Peng W., Ye Y., Rabinovich B. A., Liu C., Lou Y., Zhang M., Whittington M., Yang Y., Overwijk W. W., Lizee G., Hwu P., Transduction of tumor-specific T cells with CXCR2 chemokine receptor improves migration to tumor and antitumor immune responses. Clin. Cancer Res. 16, 5458–5468 (2010). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 99.Moon E. K., Carpenito C., Sun J., Wang L. C., Kapoor V., Predina J., Powell D. J. Jr., Riley J. L., June C. H., Albelda S. M., 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. 17, 4719–4730 (2011). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 100.Park J. S., Rhau B., Hermann A., McNally K. A., Zhou C., Gong D., Weiner O. D., Conklin B. R., Onuffer J., Lim W. A., Synthetic control of mammalian-cell motility by engineering chemotaxis to an orthogonal bioinert chemical signal. Proc. Natl. Acad. Sci. U.S.A. 111, 5896–5901 (2014). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 101.Xu Y., Hyun Y. M., Lim K., Lee H., Cummings R. J., Gerber S. A., Bae S., Cho T. Y., Lord E. M., Kim M., Optogenetic control of chemokine receptor signal and T-cell migration. Proc. Natl. Acad. Sci. U.S.A. 111, 6371–6376 (2014). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 102.Vincent R. L., Gurbatri C. R., Li F., Vardoshvili A., Coker C., Im J., Ballister E. R., Rouanne M., Savage T., de Los Santos-Alexis K., Redenti A., Brockmann L., Komaranchath M., Arpaia N., Danino T., Probiotic-guided CAR-T cells for solid tumor targeting. Science 382, 211–218 (2023). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 103.Juillerat A., Marechal A., Filhol J. M., Valogne Y., Valton J., Duclert A., Duchateau P., Poirot L., An oxygen sensitive self-decision making engineered CAR T-cell. Sci. Rep. 7, 39833 (2017). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 104.Kosti P., Opzoomer J. W., Larios-Martinez K. I., Henley-Smith R., Scudamore C. L., Okesola M., Taher M. Y. M., Davies D. M., Muliaditan T., Larcombe-Young D., Woodman N., Gillett C. E., Thavaraj S., Maher J., Arnold J. N., Hypoxia-sensing CAR T cells provide safety and efficacy in treating solid tumors. Cell Rep. Med. 2, 100227 (2021). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 105.Singhal R., Shah Y. M., Oxygen battle in the gut: Hypoxia and hypoxia-inducible factors in metabolic and inflammatory responses in the intestine. J. Biol. Chem. 295, 10493–10505 (2020). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 106.Dumas S. J., Meta E., Borri M., Luo Y., Li X., Rabelink T. J., Carmeliet P., Phenotypic diversity and metabolic specialization of renal endothelial cells. Nat. Rev. Nephrol. 17, 441–464 (2021). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 107.Han X., Bryson P. D., Zhao Y., Cinay G. E., Li S., Guo Y., Siriwon N., Wang P., Masked chimeric antigen receptor for tumor-specific activation. Mol. Ther. 25, 274–284 (2017). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 108.Zhang B., Wang Y., Huang S., Sun J., Wang M., Ma W., You Y., Wu L., Hu J., Song W., Liu X., Li S., Chen H., Zhang G., Zhang L., Zhou D., Li L., Zhang X., Photoswitchable CAR-T cell function in vitro and in vivo via a cleavable mediator. Cell Chem. Biol. 28, 60–69.e7 (2021). [DOI] [PubMed] [Google Scholar]
- 109.Kvorjak M., Ruffo E., Tivon Y., So V., Parikh A. B., Deiters A., Lohmueller J., Conditional control of universal CAR T cells by cleavable OFF-switch adaptors. ACS Synth. Biol. 12, 2996–3007 (2023). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 110.Huang Z., Wu Y., Allen M. E., Pan Y., Kyriakakis P., Lu S., Chang Y. J., Wang X., Chien S., Wang Y., Engineering light-controllable CAR T cells for cancer immunotherapy. Sci. Adv. 6, eaay9209 (2020). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 111.Miller I. C., Zamat A., Sun L. K., Phuengkham H., Harris A. M., Gamboa L., Yang J., Murad J. P., Priceman S. J., Kwong G. A., Enhanced intratumoural activity of CAR T cells engineered to produce immunomodulators under photothermal control. Nat. Biomed. Eng. 5, 1348–1359 (2021). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 112.Wu Y., Liu Y., Huang Z., Wang X., Jin Z., Li J., Limsakul P., Zhu L., Allen M., Pan Y., Bussell R., Jacobson A., Liu T., Chien S., Wang Y., Control of the activity of CAR-T cells within tumours via focused ultrasound. Nat. Biomed. Eng. 5, 1336–1347 (2021). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 113.U.S. Food and Drug Administration, FDA Investigating Serious Risk of T-Cell Malignancy Following BCMA-Directed or CD19-Directed Autologous Chimeric Antigen Receptor (CAR) T Cell Immunotherapies (U.S. Food and Drug Administration, 2023). [Google Scholar]
- 114.Lareau C. A., Yin Y., Maurer K., Sandor K. D., Daniel B., Yagnik G., Pena J., Crawford J. C., Spanjaart A. M., Gutierrez J. C., Haradhvala N. J., Riberdy J. M., Abay T., Stickels R. R., Verboon J. M., Liu V., Buquicchio F. A., Wang F., Southard J., Song R., Li W., Shrestha A., Parida L., Getz G., Maus M. V., Li S., Moore A., Roberts Z. J., Ludwig L. S., Talleur A. C., Thomas P. G., Dehghani H., Pertel T., Kundaje A., Gottschalk S., Roth T. L., Kersten M. J., Wu C. J., Majzner R. G., Satpathy A. T., Latent human herpesvirus 6 is reactivated in CAR T cells. Nature 623, 608–615 (2023). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 115.Zhang M., Wei G., Zhou L., Zhou J., Chen S., Zhang W., Wang D., Luo X., Cui J., Huang S., Fu S., Zhou X., Tang Y., Ding X., Kuang J., He X. P., Hu Y., Huang H., GPRC5D CAR T cells (OriCAR-017) in patients with relapsed or refractory multiple myeloma (POLARIS): A first-in-human, single-centre, single-arm, phase 1 trial. Lancet Haematol. 10, e107–e116 (2023). [DOI] [PubMed] [Google Scholar]
- 116.Xia J., Li H., Yan Z., Zhou D., Wang Y., Qi Y., Cao J., Li D., Cheng H., Sang W., Zhu F., Sun H., Chen W., Qi K., Yan D., Qiu T., Qiao J., Yao R., Liu Y., Wang X., Zhang Y., Peng S., Huang C. H., Zheng J., Li Z., Chang A. H., Xu K., Anti–G protein–coupled receptor, class C group 5 member D chimeric antigen receptor T cells in patients with relapsed or refractory multiple myeloma: A single-arm, phase II trial. J. Clin. Oncol. 41, 2583–2593 (2023). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 117.Rodriguez-Otero P., Ailawadhi S., Arnulf B., Patel K., Cavo M., Nooka A. K., Manier S., Callander N., Costa L. J., Vij R., Bahlis N. J., Moreau P., Solomon S. R., Delforge M., Berdeja J., Truppel-Hartmann A., Yang Z., Favre-Kontula L., Wu F., Piasecki J., Cook M., Giralt S., Ide-cel or standard regimens in relapsed and refractory multiple myeloma. N. Engl. J. Med. 388, 1002–1014 (2023). [DOI] [PubMed] [Google Scholar]
- 118.Mailankody S., Matous J. V., Chhabra S., Liedtke M., Sidana S., Oluwole O. O., Malik S., Nath R., Anwer F., Cruz J. C., Htut M., Karski E. E., Lovelace W., Dillon M., Butz E., Ying W., Balakumaran A., Kumar S. K., Allogeneic BCMA-targeting CAR T cells in relapsed/refractory multiple myeloma: Phase 1 UNIVERSAL trial interim results. Nat. Med. 29, 422–429 (2023). [DOI] [PubMed] [Google Scholar]
- 119.Qu X., An G., Sui W., Wang T., Zhang X., Yang J., Zhang Y., Zhang L., Zhu D., Huang J., Zhu S., Yao X., Li J., Zheng C., Zhu K., Wei Y., Lv X., Lan L., Yao Y., Zhou D., Lu P., Qiu L., Li J., Phase 1 study of C-CAR088, a novel humanized anti-BCMA CAR T-cell therapy in relapsed/refractory multiple myeloma. J. Immunother. Cancer 10, e005145 (2022). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 120.Zhao W. H., Wang B. Y., Chen L. J., Fu W. J., Xu J., Liu J., Jin S. W., Chen Y. X., Cao X. M., Yang Y., Zhang Y. L., Wang F. X., Zhang P. Y., Lei B., Gu L. F., Wang J. L., Zhang H., Bai J., Xu Y., Zhu H., Du J., Jiang H., Fan X. H., Li J. Y., Hou J., Chen Z., Zhang W. G., Mi J. Q., Chen S. J., He A. L., Four-year follow-up of LCAR-B38M in relapsed or refractory multiple myeloma: A phase 1, single-arm, open-label, multicenter study in China (LEGEND-2). J. Hematol. Oncol. 15, 86 (2022). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 121.Du J., Wei R., Jiang S., Jiang H., Li L., Qiang W., He H., Shi L., Ma Q., Yu K., Zhang X., Ding H., Sun X., Xiang F., Zhu L., Cheng Z., Fu W., CAR-T cell therapy targeting B cell maturation antigen is effective for relapsed/refractory multiple myeloma, including cases with poor performance status. Am. J. Hematol. 97, 933–941 (2022). [DOI] [PubMed] [Google Scholar]
- 122.Frigault M. J., Bishop M. R., Rosenblatt J., O'Donnell E. K., Raje N., Cook D., Yee A. J., Logan E., Avigan D. E., Jakubowiak A., Shaw K., Daley H., Nikiforow S., Griffin F., Cornwell C., Shen A., Heery C., Maus M. V., Phase 1 study of CART-ddBCMA for the treatment of subjects with relapsed and refractory multiple myeloma. Blood Adv. 7, 768–777 (2023). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 123.San-Miguel J., Dhakal B., Yong K., Spencer A., Anguille S., Mateos M. V., Fernandez de Larrea C., Martinez-Lopez J., Moreau P., Touzeau C., Leleu X., Avivi I., Cavo M., Ishida T., Kim S. J., Roeloffzen W., van de Donk N., Dytfeld D., Sidana S., Costa L. J., Oriol A., Popat R., Khan A. M., Cohen Y. C., Ho P. J., Griffin J., Lendvai N., Lonardi C., Slaughter A., Schecter J. M., Jackson C. C., Connors K., Li K., Zudaire E., Chen D., Gilbert J., Yeh T. M., Nagle S., Florendo E., Pacaud L., Patel N., Harrison S. J., Einsele H., Cilta-cel or standard care in lenalidomide-refractory multiple myeloma. N. Engl. J. Med. 389, 335–347 (2023). [DOI] [PubMed] [Google Scholar]
- 124.Asherie N., Kfir-Erenfeld S., Avni B., Assayag M., Dubnikov T., Zalcman N., Lebel E., Zimran E., Shaulov A., Pick M., Cohen Y., Avivi I., Cohen C., Gatt M. E., Grisariu S., Stepensky P., Development and manufacture of novel locally produced anti-BCMA CAR T cells for the treatment of relapsed/refractory multiple myeloma: Results from a phase I clinical trial. Haematologica 108, 1827–1839 (2023). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 125.Cowan A. J., Pont M. J., Sather B. D., Turtle C. J., Till B. G., Libby E. N. III, Coffey D. G., Tuazon S. A., Wood B., Gooley T., Wu V. Q., Voutsinas J., Song X., Shadman M., Gauthier J., Chapuis A. G., Milano F., Maloney D. G., Riddell S. R., Green D. J., γ-Secretase inhibitor in combination with BCMA chimeric antigen receptor T-cell immunotherapy for individuals with relapsed or refractory multiple myeloma: A phase 1, first-in-human trial. Lancet Oncol. 24, 811–822 (2023). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 126.Minakata D., Ishida T., Ando K., Suzuki R., Tanaka J., Hagiwara S., Ananthakrishnan R., Kuwayama S., Nishio M., Kanda Y., Suzuki K., Phase 2 results of idecabtagene vicleucel (ide-cel, bb2121) in Japanese patients with relapsed and refractory multiple myeloma. Int. J. Hematol. 117, 729–737 (2023). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 127.Lin Y., Raje N. S., Berdeja J. G., Siegel D. S., Jagannath S., Madduri D., Liedtke M., Rosenblatt J., Maus M. V., Massaro M., Petrocca F., Yeri A., Finney O., Caia A., Yang Z., Martin N., Campbell T. B., Rytlewski J., Fuller J., Hege K., Munshi N. C., Kochenderfer J. N., Idecabtagene vicleucel for relapsed and refractory multiple myeloma: Post hoc 18-month follow-up of a phase 1 trial. Nat. Med. 29, 2286–2294 (2023). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 128.Niu J., Qiu H., Xiang F., Zhu L., Yang J., Huang C., Zhou K., Tong Y., Cai Y., Dong B., Lu Y., Sun X., Wan L., Ding X., Wang H., Song X., CD19/CD22 bispecific CAR-T cells for MRD-positive adult B cell acute lymphoblastic leukemia: A phase I clinical study. Blood Cancer J. 13, 44 (2023). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 129.Shalabi H., Qin H., Su A., Yates B., Wolters P. L., Steinberg S. M., Ligon J. A., Silbert S., DeDe K., Benzaoui M., Goldberg S., Achar S., Schneider D., Shahani S. A., Little L., Foley T., Molina J. C., Panch S., Mackall C. L., Lee D. W., Chien C. D., Pouzolles M., Ahlman M., Yuan C. M., Wang H. W., Wang Y., Inglefield J., Toledo-Tamula M. A., Martin S., Highfill S. L., Altan-Bonnet G., Stroncek D., Fry T. J., Taylor N., Shah N. N., CD19/22 CAR T cells in children and young adults with B-ALL: Phase 1 results and development of a novel bicistronic CAR. Blood 140, 451–463 (2022). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 130.Yang J., He J., Zhang X., Li J., Wang Z., Zhang Y., Qiu L., Wu Q., Sun Z., Ye X., Yin W., Cao W., Shen L., Sersch M., Lu P., Next-day manufacture of a novel anti-CD19 CAR-T therapy for B-cell acute lymphoblastic leukemia: First-in-human clinical study. Blood Cancer J. 12, 104 (2022). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 131.Talleur A. C., Qudeimat A., Metais J. Y., Langfitt D., Mamcarz E., Crawford J. C., Huang S., Cheng C., Hurley C., Madden R., Sharma A., Suliman A., Srinivasan A., Velasquez M. P., Obeng E. A., Willis C., Akel S., Karol S. E., Inaba H., Bragg A., Zheng W., Zhou S. M., Schell S., Tuggle-Brown M., Cullins D., Patil S. L., Li Y., Thomas P. G., Zebley C., Youngblood B., Pui C. H., Lockey T., Geiger T. L., Meagher M. M., Triplett B. M., Gottschalk S., Preferential expansion of CD8+ CD19-CAR T cells postinfusion and the role of disease burden on outcome in pediatric B-ALL. Blood Adv. 6, 5737–5749 (2022). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 132.Qi Y., Zhao M., Hu Y., Wang Y., Li P., Cao J., Shi M., Tan J., Zhang M., Xiao X., Xia J., Ma S., Qiao J., Yan Z., Li H., Pan B., Sang W., Li D., Li Z., Zhou J., Huang H., Liang A., Zheng J., Xu K., Efficacy and safety of CD19-specific CAR T cell-based therapy in B-cell acute lymphoblastic leukemia patients with CNSL. Blood 139, 3376–3386 (2022). [DOI] [PubMed] [Google Scholar]
- 133.Frigault M. J., Dietrich J., Gallagher K., Roschewski M., Jordan J. T., Forst D., Plotkin S. R., Cook D., Casey K. S., Lindell K. A., Depinho G. D., Katsis K., Elder E. L., Leick M. B., Choi B., Horick N., Preffer F., Saylor M., McAfee S., O'Donnell P. V., Spitzer T. R., Dey B., DeFilipp Z., El-Jawahri A., Batchelor T. T., Maus M. V., Chen Y. B., Safety and efficacy of tisagenlecleucel in primary CNS lymphoma: A phase 1/2 clinical trial. Blood 139, 2306–2315 (2022). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 134.Hu Y., Zhou Y., Zhang M., Zhao H., Wei G., Ge W., Cui Q., Mu Q., Chen G., Han L., Guo T., Cui J., Jiang X., Zheng X., Yu S., Li X., Zhang X., Chen M., Li X., Gao M., Wang K., Zu C., Zhang H., He X., Wang Y., Wang D., Ren J., Huang H., Genetically modified CD7-targeting allogeneic CAR-T cell therapy with enhanced efficacy for relapsed/refractory CD7-positive hematological malignancies: A phase I clinical study. Cell Res. 32, 995–1007 (2022). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 135.Chiesa R., Georgiadis C., Syed F., Zhan H., Etuk A., Gkazi S. A., Preece R., Ottaviano G., Braybrook T., Chu J., Kubat A., Adams S., Thomas R., Gilmour K., O'Connor D., Vora A., Qasim W., Base-Edited C. A. R. T. G., Base-edited CAR7 T cells for relapsed T-cell acute lymphoblastic leukemia. N. Engl. J. Med. 389, 899–910 (2023). [DOI] [PubMed] [Google Scholar]
- 136.Pan J., Tang K., Luo Y., Seery S., Tan Y., Deng B., Liu F., Xu X., Ling Z., Song W., Xu J., Duan J., Wang Z., Li C., Wang K., Zhang Y., Yu X., Zheng Q., Zhao L., Zhang J., Chang A. H., Feng X., Sequential CD19 and CD22 chimeric antigen receptor T-cell therapy for childhood refractory or relapsed B-cell acute lymphocytic leukaemia: A single-arm, phase 2 study. Lancet Oncol. 24, 1229–1241 (2023). [DOI] [PubMed] [Google Scholar]
- 137.Sehgal A., Hoda D., Riedell P. A., Ghosh N., Hamadani M., Hildebrandt G. C., Godwin J. E., Reagan P. M., Wagner-Johnston N., Essell J., Nath R., Solomon S. R., Champion R., Licitra E., Fanning S., Gupta N., Dubowy R., D'Andrea A., Wang L., Ogasawara K., Thorpe J., Gordon L. I., Lisocabtagene maraleucel as second-line therapy in adults with relapsed or refractory large B-cell lymphoma who were not intended for haematopoietic stem cell transplantation (PILOT): An open-label, phase 2 study. Lancet Oncol. 23, 1066–1077 (2022). [DOI] [PubMed] [Google Scholar]
- 138.Kamdar M., Solomon S. R., Arnason J., Johnston P. B., Glass B., Bachanova V., Ibrahimi S., Mielke S., Mutsaers P., Hernandez-Ilizaliturri F., Izutsu K., Morschhauser F., Lunning M., Maloney D. G., Crotta A., Montheard S., Previtali A., Stepan L., Ogasawara K., Mack T., Abramson J. S., Investigators T., Lisocabtagene maraleucel versus standard of care with salvage chemotherapy followed by autologous stem cell transplantation as second-line treatment in patients with relapsed or refractory large B-cell lymphoma (TRANSFORM): Results from an interim analysis of an open-label, randomised, phase 3 trial. Lancet 399, 2294–2308 (2022). [DOI] [PubMed] [Google Scholar]
- 139.Makita S., Yamamoto G., Maruyama D., Asano-Mori Y., Kaji D., Ananthakrishnan R., Ogasawara K., Stepan L., Schusterbauer C., Rettby N., Hasskarl J., Izutsu K., Phase 2 results of lisocabtagene maraleucel in Japanese patients with relapsed/refractory aggressive B-cell non-Hodgkin lymphoma. Cancer Med. 11, 4889–4899 (2022). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 140.Neelapu S. S., Dickinson M., Munoz J., Ulrickson M. L., Thieblemont C., Oluwole O. O., Herrera A. F., Ujjani C. S., Lin Y., Riedell P. A., Kekre N., de Vos S., Lui C., Milletti F., Dong J., Xu H., Chavez J. C., Axicabtagene ciloleucel as first-line therapy in high-risk large B-cell lymphoma: The phase 2 ZUMA-12 trial. Nat. Med. 28, 735–742 (2022). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 141.Jacobson C. A., Rosenthal A. C., Arnason J., Agarwal S., Zhang P., Wu W., Amber V., Yared J. A., A phase 2 trial of defibrotide for the prevention of chimeric antigen receptor T-cell-associated neurotoxicity syndrome. Blood Adv. 7, 6790–6799 (2023). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 142.Jaeger U., Worel N., McGuirk J. P., Riedell P. A., Fleury I., Du Y., Han X., Pearson D., Redondo S., Waller E. K., Safety and efficacy of tisagenlecleucel plus pembrolizumab in patients with r/r DLBCL: Phase 1b PORTIA study results. Blood Adv. 7, 2283–2286 (2023). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 143.Ying Z., Yang H., Guo Y., Li W., Zou D., Zhou D., Wang Z., Zhang M., Wu J., Liu H., Wang C., Ma L., Yang S., Zhou Z., Qin Y., Song Y., Zhu J., Long-term outcomes of relmacabtagene autoleucel in Chinese patients with relapsed/refractory large B-cell lymphoma: Updated results of the RELIANCE study. Cytotherapy 25, 521–529 (2023). [DOI] [PubMed] [Google Scholar]
- 144.Zhang Y., Wang Y., Liu Y., Tong C., Wang C., Guo Y., Ti D., Yang Q., Qiao S., Wu Z., Han W., Long-term activity of tandem CD19/CD20 CAR therapy in refractory/relapsed B-cell lymphoma: A single-arm, phase 1-2 trial. Leukemia 36, 189–196 (2022). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 145.Ying Z., Xie Y., Zheng W., Liu W., Lin N., Tu M., Wang X., Ping L., Deng L., Zhang C., Wu M., Feng F., Du T., Tang Y., Su F., Guo Z., Li J., Song Y., Zhu J., Efficacy and safety of relmacabtagene autoleucel, an anti-CD19 chimeric antigen receptor T cell, in relapsed/refractory B-cell non-Hodgkin's lymphoma: 2-year results of a phase 1 trial. Bone Marrow Transplant. 58, 288–294 (2023). [DOI] [PubMed] [Google Scholar]
- 146.Singh H., Srour S. A., Milton D. R., McCarty J., Dai C., Gaballa M. R., Ammari M., Olivares S., Huls H., De Groot E., Marin D., Petropoulos D., Olson A. L., Anderlini P., Im J. S., Khouri I., Hosing C. M., Rezvani K., Champlin R. E., Shpall E. J., Cooper L. J. N., Kebriaei P., Sleeping beauty generated CD19 CAR T-cell therapy for advanced B-cell hematological malignancies. Front. Immunol. 13, 1032397 (2022). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 147.Erdogan E., Yalcin K., Hemsinlioglu C., Sezgin A., Seyis U., Kancagi D. D., Tastan C., Yurtsever B., Turan R. D., Cakirsoy D., Abanuz S., Karakus G. S., Elek M., Bekoz H. S., Gemici A. I., Sargin D., Arat M., Ferhanoglu B., Pekguc E., Ornek S., Buyuktas D., Birgen N., Ratip S., Ovali E., Preliminary report of the academic CAR-T (ISIKOK-19) cell clinical trial in Turkey: Characterization of product and outcomes of clinical application. Turk. J. Haematol. 39, 206–210 (2022). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 148.Lu P., Liu Y., Yang J., Zhang X., Yang X., Wang H., Wang L., Wang Q., Jin D., Li J., Huang X., Naturally selected CD7 CAR-T therapy without genetic manipulations for T-ALL/LBL: First-in-human phase 1 clinical trial. Blood 140, 321–334 (2022). [DOI] [PubMed] [Google Scholar]
- 149.Zhang M., Chen D., Fu X., Meng H., Nan F., Sun Z., Yu H., Zhang L., Li L., Li X., Wang X., Wang M., You F., Li Z., Chang Y., Zhou Z., Yan J., Li J., Wu X., Wang Y., Wang Y., Xiang S., Chen Y., Pan G., Xu H., Zhang B., Yang L., Autologous nanobody-derived fratricide-resistant CD7-CAR T-cell therapy for patients with relapsed and refractory T-cell acute lymphoblastic leukemia/lymphoma. Clin. Cancer Res. 28, 2830–2843 (2022). [DOI] [PubMed] [Google Scholar]
- 150.Qin C., Tian D. S., Zhou L. Q., Shang K., Huang L., Dong M. H., You Y. F., Xiao J., Xiong Y., Wang W., Pang H., Guo J. J., Cai S. B., Wang D., Li C. R., Zhang M., Bu B. T., Wang W., Anti-BCMA CAR T-cell therapy CT103A in relapsed or refractory AQP4-IgG seropositive neuromyelitis optica spectrum disorders: Phase 1 trial interim results. Signal Transduct. Target. Ther. 8, 5 (2023). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 151.Qi C., Gong J., Li J., Liu D., Qin Y., Ge S., Zhang M., Peng Z., Zhou J., Cao Y., Zhang X., Lu Z., Lu M., Yuan J., Wang Z., Wang Y., Peng X., Gao H., Liu Z., Wang H., Yuan D., Xiao J., Ma H., Wang W., Li Z., Shen L., Claudin18.2-specific CAR T cells in gastrointestinal cancers: Phase 1 trial interim results. Nat. Med. 28, 1189–1198 (2022). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 152.Narayan V., Barber-Rotenberg J. S., Jung I. Y., Lacey S. F., Rech A. J., Davis M. M., Hwang W. T., Lal P., Carpenter E. L., Maude S. L., Plesa G., Vapiwala N., Chew A., Moniak M., Sebro R. A., Farwell M. D., Marshall A., Gilmore J., Lledo L., Dengel K., Church S. E., Hether T. D., Xu J., Gohil M., Buckingham T. H., Yee S. S., Gonzalez V. E., Kulikovskaya I., Chen F., Tian L., Tien K., Gladney W., Nobles C. L., Raymond H. E., Program I. P. C. C. T., Hexner E. O., Siegel D. L., Bushman F. D., June C. H., Fraietta J. A., Haas N. B., PSMA-targeting TGFbeta-insensitive armored CAR T cells in metastatic castration-resistant prostate cancer: A phase 1 trial. Nat. Med. 28, 724–734 (2022). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 153.Mackensen A., Haanen J., Koenecke C., Alsdorf W., Wagner-Drouet E., Borchmann P., Heudobler D., Ferstl B., Klobuch S., Bokemeyer C., Desuki A., Luke F., Kutsch N., Muller F., Smit E., Hillemanns P., Karagiannis P., Wiegert E., He Y., Ho T., Kang-Fortner Q., Schlitter A. M., Schulz-Eying C., Finlayson A., Flemmig C., Kuhlcke K., Preussner L., Rengstl B., Tureci O., Sahin U., CLDN6-specific CAR-T cells plus amplifying RNA vaccine in relapsed or refractory solid tumors: The phase 1 BNT211-01 trial. Nat. Med. 29, 2844–2853 (2023). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 154.Shah P. D., Huang A. C., Xu X., Orlowski R., Amaravadi R. K., Schuchter L. M., Zhang P., Tchou J., Matlawski T., Cervini A., Shea J., Gilmore J., Lledo L., Dengel K., Marshall A., Wherry E. J., Linette G. P., Brennan A., Gonzalez V., Kulikovskaya I., Lacey S. F., Plesa G., June C. H., Vonderheide R. H., Mitchell T. C., Phase I trial of autologous RNA-electroporated cMET-directed CAR T cells administered intravenously in patients with melanoma and breast carcinoma. Cancer Res. Commun. 3, 821–829 (2023). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 155.Li D., Guo X., Yang K., Yang Y., Zhou W., Huang Y., Liang X., Su J., Jiang L., Li J., Fu M., He H., Yang J., Shi H., Yang H., Tong A., Chen N., Hu J., Xu Q., Wei Y. Q., Wang W., EpCAM-targeting CAR-T cell immunotherapy is safe and efficacious for epithelial tumors. Sci. Adv. 9, eadg9721 (2023). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 156.Granit V., Benatar M., Kurtoglu M., Miljkovic M. D., Chahin N., Sahagian G., Feinberg M. H., Slansky A., Vu T., Jewell C. M., Singer M. S., Kalayoglu M. V., Howard J. F. Jr., Mozaffar T., MG-001 Study Team , Safety and clinical activity of autologous RNA chimeric antigen receptor T-cell therapy in myasthenia gravis (MG-001): A prospective, multicentre, open-label, non-randomised phase 1b/2a study. Lancet Neurol. 22, 578–590 (2023). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 157.Morgan R. A., Yang J. C., Kitano M., Dudley M. E., Laurencot C. M., Rosenberg S. A., Case report of a serious adverse event following the administration of T cells transduced with a chimeric antigen receptor recognizing ERBB2. Mol. Ther. 18, 843–851 (2010). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 158.Thistlethwaite F. C., Gilham D. E., Guest R. D., Rothwell D. G., Pillai M., Burt D. J., Byatte A. J., Kirillova N., Valle J. W., Sharma S. K., Chester K. A., Westwood N. B., Halford S. E. R., Nabarro S., Wan S., Austin E., Hawkins R. E., The clinical efficacy of first-generation carcinoembryonic antigen (CEACAM5)-specific CAR T cells is limited by poor persistence and transient pre-conditioning-dependent respiratory toxicity. Cancer Immunol. Immunother. 66, 1425–1436 (2017). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 159.Lamers C. H., Sleijfer S., van Steenbergen S., van Elzakker P., van Krimpen B., Groot C., Vulto A., den Bakker M., Oosterwijk E., Debets R., Gratama J. W., Treatment of metastatic renal cell carcinoma with CAIX CAR-engineered T cells: Clinical evaluation and management of on-target toxicity. Mol. Ther. 21, 904–912 (2013). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 160.Dai H., Tong C., Shi D., Chen M., Guo Y., Chen D., Han X., Wang H., Wang Y., Shen P., Efficacy and biomarker analysis of CD133-directed CAR T cells in advanced hepatocellular carcinoma: A single-arm, open-label, phase II trial. Onco. Targets. Ther. 9, 1846926 (2020). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 161.Kochenderfer J. N., Dudley M. E., Feldman S. A., Wilson W. H., Spaner D. E., Maric I., Stetler-Stevenson M., Phan G. Q., Hughes M. S., Sherry R. M., Yang J. C., Kammula U. S., Devillier L., Carpenter R., Nathan D. A., Morgan R. A., Laurencot C., Rosenberg S. A., B-cell depletion and remissions of malignancy along with cytokine-associated toxicity in a clinical trial of anti-CD19 chimeric-antigen-receptor-transduced T cells. Blood 119, 2709–2720 (2012). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 162.Foster M. C., Savoldo B., Lau W., Rubinos C., Grover N., Armistead P., Coghill J., Hagan R. S., Morrison K., Buchanan F. B., Cheng C., Laing S., Ivanova A., West J., Foster A., Serody J., Dotti G., Utility of a safety switch to abrogate CD19.CAR T-cell-associated neurotoxicity. Blood 137, 3306–3309 (2021). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 163.Liu Z., Zhou J., Yang X., Liu Y., Zou C., Lv W., Chen C., Cheng K. K., Chen T., Chang L. J., Wu D., Mao J., Safety and antitumor activity of GD2-specific 4SCAR-T cells in patients with glioblastoma. Mol. Cancer 22, 3 (2023). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 164.Majzner R. G., Ramakrishna S., Yeom K. W., Patel S., Chinnasamy H., Schultz L. M., Richards R. M., Jiang L., Barsan V., Mancusi R., Geraghty A. C., Good Z., Mochizuki A. Y., Gillespie S. M., Toland A. M. S., Mahdi J., Reschke A., Nie E. H., Chau I. J., Rotiroti M. C., Mount C. W., Baggott C., Mavroukakis S., Egeler E., Moon J., Erickson C., Green S., Kunicki M., Fujimoto M., Ehlinger Z., Reynolds W., Kurra S., Warren K. E., Prabhu S., Vogel H., Rasmussen L., Cornell T. T., Partap S., Fisher P. G., Campen C. J., Filbin M. G., Grant G., Sahaf B., Davis K. L., Feldman S. A., Mackall C. L., Monje M., GD2-CAR T cell therapy for H3K27M-mutated diffuse midline gliomas. Nature 603, 934–941 (2022). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 165.Heczey A., Louis C. U., Savoldo B., Dakhova O., Durett A., Grilley B., Liu H., Wu M. F., Mei Z., Gee A., Mehta B., Zhang H., Mahmood N., Tashiro H., Heslop H. E., Dotti G., Rooney C. M., Brenner M. K., CAR T cells administered in combination with lymphodepletion and PD-1 inhibition to patients with neuroblastoma. Mol. Ther. 25, 2214–2224 (2017). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 166.Ghosn M., Cheema W., Zhu A., Livschitz J., Maybody M., Boas F. E., Santos E., Kim D., Beattie J. A., Offin M., Rusch V. W., Zauderer M. G., Adusumilli P. S., Solomon S. B., Image-guided interventional radiological delivery of chimeric antigen receptor (CAR) T cells for pleural malignancies in a phase I/II clinical trial. Lung Cancer 165, 1–9 (2022). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 167.Ceppi F., Rivers J., Annesley C., Pinto N., Park J. R., Lindgren C., Mgebroff S., Linn N., Delaney M., Gardner R. A., Lymphocyte apheresis for chimeric antigen receptor T-cell manufacturing in children and young adults with leukemia and neuroblastoma. Transfusion 58, 1414–1420 (2018). [DOI] [PubMed] [Google Scholar]
- 168.Koneru M., O'Cearbhaill R., Pendharkar S., Spriggs D. R., Brentjens R. J., A phase I clinical trial of adoptive T cell therapy using IL-12 secreting MUC-16ecto directed chimeric antigen receptors for recurrent ovarian cancer. J. Transl. Med. 13, 102 (2015). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 169.Park J. R., Digiusto D. L., Slovak M., Wright C., Naranjo A., Wagner J., Meechoovet H. B., Bautista C., Chang W. C., Ostberg J. R., Jensen M. C., Adoptive transfer of chimeric antigen receptor re-directed cytolytic T lymphocyte clones in patients with neuroblastoma. Mol. Ther. 15, 825–833 (2007). [DOI] [PubMed] [Google Scholar]
- 170.Brown C. E., Badie B., Barish M. E., Weng L., Ostberg J. R., Chang W. C., Naranjo A., Starr R., Wagner J., Wright C., Zhai Y., Bading J. R., Ressler J. A., Portnow J., D'Apuzzo M., Forman S. J., Jensen M. C., Bioactivity and safety of IL13Rα2-redirected chimeric antigen receptor CD8+ T cells in patients with recurrent glioblastoma. Clin. Cancer Res. 21, 4062–4072 (2015). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 171.Stornaiuolo A., Valentinis B., Sirini C., Scavullo C., Asperti C., Zhou D., Martinez De La Torre Y., Corna S., Casucci M., Porcellini S., Traversari C., Characterization and functional analysis of CD44v6.CAR T cells endowed with a new low-affinity nerve growth factor receptor-based spacer. Hum. Gene Ther. 32, 744–760 (2021). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 172.Chang A., Phillips S., Lee M., Huang E., Anand N., Ko H., Tran S., Rios L., Dai R., Nguyen A., Vasudevan A., Steinfeld R. H., Polyak D., Fuhriman J., Yun H., Feng J., Wang H., Tran B., Smith J., Sirichoke T., 277 Development of a robust manufacturing process for AB-1015, an integrated circuit T cell (ICT) product, using targeted, CRISPR integration of transgenes by electroporation (CITE) editing. J. Immunother. Cancer 10, A292 (2022). [Google Scholar]
- 173.Pampusch M. S., Skinner P. J., Transduction and expansion of primary T cells in nine days with maintenance of central memory phenotype. J. Vis. Exp. 157, e60400 (2020). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 174. EXUMA Biotechnology and affiliate Shanghai PerHum Therapeutics, Shanghai Perhum Therapeutics announces preliminary results of two first-in-human solid tumor CAR-T products. 21 May 2019. https://exumabio.com/news/2019/05/shanghai-perhum-therapeutics-announces-preliminary-results-of-two-first-in-human-solid-tumor-car-t-products/.



