Abstract
Chimeric antigen receptor T (CAR-T) cell therapy is an innovative and promising approach to treat cancer. Clinical trials have demonstrated remarkable results, providing hope for patients who have exhausted more traditional therapies. However, this new therapy is not without challenges, as significant side effects have been associated with it. Cytokine release syndrome (CRS) is a widely recognized and consequential side effect of CAR-T cell therapy. Neurological toxicity is another potential side effect that can cause confusion and seizures in some patients. Hematologic toxicities, such as anemia and thrombocytopenia, can increase the risk of bleeding or infection. B-cell aplasia can also occur, leading to increased vulnerability to infections. Strategies to reduce the incidence and severity of toxicities include suicide, endogenous, and exogenous switches to modulate the activity of the immune system toward cancer while minimizing toxicity. Despite the obstacles faced by CAR-T cell therapy, continuous research and development in this area offer considerable potential for improving this treatment as a more reliable and efficient method for treating cancer.
Keywords: CAR-T cell, cell therapy, challenge, immunotherapy, side effects
INTRODUCTION
Targeted immunotherapy is considered one of the optimal cancer therapies among various treatment methods.[1] This treatment is achieved when tumor cells are eradicated without damaging healthy tissue or causing the toxicity associated with conventional chemotherapy.[2,3] The history of producing these cells dates back to less than two decades ago, when numerous influential monoclonal antibodies were introduced to treat cancer.[4] Despite the remarkable success of immunotherapy using monoclonal antibodies against specific cancer antigens, there is a short period of stability for these antibodies in the patient’s bloodstream.[5] These cells have proven successful and promising in clinical studies, particularly when used in lymphoblastic leukemia and Hodgkin’s lymphoma.[6,7] They have armed T lymphocytes with a chimeric antigen receptor (CAR), similar to the active part of the antibody, by modeling the amino acid structure of the monoclonal antibody.[8] These cells possess both the abilities of T lymphocytes and the specificity of monoclonal antibodies to kill target cells.[9,10] Recently, studies have expanded CAR T cell therapy beyond cancer to include cardiometabolic disorders, autoimmune diseases, fibrosis, and cellular senescence. Infectious diseases, once early targets, are also being revisited with improved understanding of immune responses and enhanced techniques, such as advanced CAR constructs, better manufacturing, and targeting various immune cell subsets like Tregs and macrophages.
While CAR-T cell therapy was first proposed in the late 1980s, it took several decades of research and development before the first generation of CAR-T cells was successfully used in clinical trials in the early 2000s.[11,12] To put it differently, the CAR-T cells of the first generation were created by genetically modifying T cells. The purpose of this modification was to enable these cells to display a specialized receptor known as CAR on their surface. The CARs were designed to selectively recognize a unique protein present only in cancer cells.[13] The second and third generations of CAR-T cells were designed with additional co-stimulatory molecules to enhance their activity and prolong their survival in the body.[14] The fourth generation of CAR-T cells, also known as TRUCKs (T-cells redirected for universal cytokine killing), is designed to secrete cytokines or other molecules that can stimulate the immune system and enhance the anti-tumor response. To accomplish this, the cells are modified to contain a section that is responsive to the nuclear factor of activated T-cells (NFAT), which includes a transgenic cytokine like IL-12.[15] When the CD3ζ-containing CARs interact with their designated target, the transgene is activated. To engineer TRUCK CAR-T cells, two transgenic cassettes are typically transferred: one for the CAR structure and another for the inducible cytokine.[16] Currently, the fifth generation of CAR-T cells is still in the early stages of development. Proposed modifications include the incorporation of gene editing techniques like CRISPR to improve the precision and specificity of CAR-T cells, as well as the use of synthetic biology techniques to create more complex cellular circuits that can respond to multiple signals in the tumor microenvironment.[17] Ongoing investigations and advancements in this area of research could lead to additional enhancements in the efficacy of CAR-T cell treatment for cancer. This type of therapy offers several advantages that make it an appealing treatment, including the ability to identify target cells independent of the human leukocyte antigens (HLA) system, kill cancer cells that utilize tumor escape mechanisms, produce large populations of tumor-specific T cells in patients, and minimally damage healthy tissues and organs.[18,19] More importantly, shorter treatment times and fewer side effects are significant achievements of CAR-T cell therapy. While these cells are promising, significant challenges remain concerning their safety and efficacy, despite the encouraging results seen in clinical trials. Specifically, while CAR-T cells have demonstrated impressive anti-tumor activity, most clinical responses have been associated with toxicity, and some patients have even experienced fatal complications following treatment.[20] In light of these concerns, a review article was recently published that aimed to survey CAR-T cell-related toxicities and explore both traditional and new methods to overcome these barriers. We emphasized the importance of understanding the mechanisms underlying CAR-T cell toxicity, as this knowledge is essential for developing effective interventions to mitigate adverse effects.
SIDE EFFECTS OF CAR-T CELL THERAPY
The therapeutic benefits of CAR-T cell therapy are often accompanied by toxic side effects, some of which can be life-threatening.[21] These toxicities can affect the cardiovascular, respiratory, and neurological systems and may manifest in various ways, such as cytokine release syndrome (CRS), tumor lysis syndrome (TLS), B-cell aplasia, graft-versus-host disease (GVHD), uncontrolled proliferation of CAR-T cells, genotoxicity, and neurotoxicity, among others. It is important to carefully monitor patients undergoing CAR-T cell therapy for potential side effects and manage them appropriately to minimize their impact on patient outcomes.[22,23] In this section, we outline and examine the potential side effects that may arise as a result of this novel approach to cancer treatment.
CRS
Immunotherapy aims to activate effector T cells and stimulate them to produce cytokines. Excessive cytokine release can lead to CRS, which varies in severity from mild to severe and can even be fatal.[24] The most commonly observed negative outcome following the administration of CAR-T cells is CRS.[25] CRS can cause a range of symptoms, including high fever, fatigue, nausea, and cardiovascular issues like tachycardia and hypotension.[26,27] In severe cases, CRS can lead to capillary leak, hepatic failure, disseminated intravascular coagulation, and other serious complications. The hallmark indicators of this syndrome typically involve elevated concentrations of certain cytokines in the body, including interferon-gamma (IFN-γ), interleukin-10, IL-6, and granulocyte-macrophage colony-stimulating factor (GM-CSF).[28,29,30,31] Notably, patients may still respond to treatment even if they do not experience CRS, particularly in the case of hematologic malignancies.[32] However, most patients with these types of cancer will experience at least mild CRS symptoms, such as fever, after receiving CAR-T cells.[33]
Following a diagnosis of CRS, identifying appropriate medications to alleviate the physiological signs of uncontrolled inflammation while preserving the anti-cancer potency of the modified cells has been challenging.[34] Systemic corticosteroids have been shown to quickly relieve CRS symptoms without jeopardizing the anti-tumor response.[31,35] However, prolonged use of high doses of corticosteroids (for over 14 days) has been linked to the depletion of the CAR-T cells transferred through adaptive means, potentially reducing their long-term anti-leukemia efficacy.[35]
The FDA has approved tocilizumab as an effective treatment for CRS associated with CAR-T cell therapy. This recombinant humanized monoclonal antibody works by blocking the activity of the IL-6 receptor (IL-6R), which is involved in the inflammatory response leading to CRS. Clinical studies have demonstrated that tocilizumab can result in near-immediate reversal of CRS symptoms in patients undergoing CAR-T cell therapy, and it is now widely used as a first-line treatment for this complication.[31,36]
TLS
This syndrome is a potential complication of CAR-T cell therapy that can arise from the breakdown of dying cancer cells.[37] When cancer cells rapidly break down in the body; uric acid, potassium, and phosphorus levels rise quicker than the kidneys can eliminate them.[38] Excess phosphorus in the body can lead to calcium absorption and lower calcium levels in the blood.[39] Changes in blood levels of these factors can affect the functioning of several organs, including the kidneys, heart, brain, muscles, and gastrointestinal tract.[38] It is important to carefully observe and evaluate patients before and after chemotherapy and CAR-T cell injections.[40] Hyperuricemia, hyperkalemia, hyperphosphatemia, and hypocalcemia should be monitored in the blood.[41] Patients should be kept hydrated, and if symptoms indicating the onset of this syndrome develop, they should be treated with allopurinol or rasburicase.[42]
B-cell aplasia
On-target off-tumor toxicity (OTOTT) is an adverse effect caused by the presence of tumor-specific antigens outside of tumor cells.[43] This occurs because CAR-T cells, which are designed to recognize and attack tumor cells, can also target normal cells during the treatment process.[44] CD19, a biomarker for the B-cell lineage, is commonly found on the surface of tumor cells in patients with hematologic malignancies.[45] The use of Anti-CD19 CAR-T cell therapy has demonstrated remarkable success in treating these types of cancers. However, it may lead to the development of B-cell aplasia since the normal B-cells, which also express CD19, can be destroyed during the treatment process.[46] Consequently, treatment can lead to the temporary or permanent loss of B lymphocytes, which the patient may need to receive immunoglobulin periodically.[47]
GVHD
It is a severe and life-threatening complication that requires prompt diagnosis and management.[48] Many patients have received CAR-T cell therapy following allo- hematopoietic stem cell transplantation (HSCT), in which the infused CAR-T cells were from a donor.[49] Despite concerns regarding alloreactivity, studies have shown that CAR-T cells collected after allo-HSCT do not cause GVHD.[29,30,31] When managing this complication, it is important to consider that immunosuppression can also affect the effectiveness and efficiency of CAR-T cells.[50]
Uncontrolled proliferation
CAR-T cells can proliferate without being regulated by normal control mechanisms.[51] Preclinical studies have shown that CAR-T cells proliferate only in response to physiological signals when exposed to targeted antigens.[52] Nevertheless, T cells can proliferate in response to signals received from malignant tumors or normal B cells, which can be beneficial or harmful depending on the intensity of that proliferation.[53] While CAR-T cell proliferation has been shown to help reduce tumor burden, uncontrolled growth can lead to the excessive accumulation of these cells within the body. In such cases, corticosteroids may be administered to manage and regulate the growth of CAR-T cells. This treatment approach helps prevent potential complications that may arise from uncontrolled growth, ensuring that the patient remains safe throughout the treatment process.[54] Overall, the uncontrolled proliferation of these cells is a potential complication of CAR-T cell therapy. However, preclinical studies have provided insights into the mechanisms behind this proliferation, and strategies are being developed to control it. With continued research, CAR-T cell therapy has the potential to become a powerful tool in the fight against cancer.
Genotoxicity
People who receive genetically engineered products are at risk for genetic modification. This risk is primarily due to viral vectors inserted into the host genome.[55] For example, following the receipt of genetically engineered products and the insertion of a transgene into the genome, a normally functioning gene may be disrupted or a proto-oncogene may be activated, or the regulatory signals may change abnormally.[56] Insertion of retroviral vectors in the vicinity of the LMO-2 oncogene has been associated with most instances of transformation.[57] Moreover, inserting a transgene into T-cells that have already differentiated may heighten the risk of inducing malignant transformation.[58] Nonetheless, so far, there haven’t been any cases where modified T-cell infusion has caused transformation. It’s worth mentioning that the LMO-2 oncogene is ordinarily inactive in T cells, making it improbable for retroviral integration to occur at this location. The use of genetically modified T cells has been shown to be safe for over ten years, without any indication of vector-induced immortalization, clonal expansion, or a preference for integration sites situated close to genes known to regulate growth or cause transformation.[59] Although the likelihood of oncogenesis resulting from gene alterations in T cells appears to be low, researchers should remain vigilant and consider this possibility.[60]
Neurotoxicity
CAR-T cell treatments can cause neurotoxicity, a severe side effect. Many patients treated with CAR-T cells develop acute neurological signs or symptoms, such as headache, confusion, delirium, language disturbances, seizures, and, in rare cases, severe cerebral edema, which are some of the clinical signs.[61] Regarding the activation and growth of CAR-T cells within a living organism, there is a connection between adverse effects on the nervous system and the release of cytokines, known as CRS.[62] Although the specific mechanisms responsible for this neurological harm are not yet understood, evidence obtained from both animals and humans indicates that the breakdown of the blood-brain barrier is implicated. An increase in cytokine levels present in the blood and cerebrospinal fluid, coupled with elevated activity in endothelial cells, has been linked to this deterioration. Patients experiencing neurotoxicity typically receive treatment comprising corticosteroids, IL-6-targeted therapies, and supportive care. However, there isn’t enough high-quality evidence to confirm their effectiveness.[63]
STRATEGIES FOR DEALING WITH TOXICITY
CAR-T cell therapy has made significant strides in treating cancer in humans, but it has also been linked to certain harmful effects and distinct toxicities.[64] Traditional anti-cancer therapies do not address the mentioned issues. For this reason, researchers have proposed innovative new approaches to reduce the incidence of toxicities and the severity of their effects. CAR-T cell therapy has been implemented to overcome adverse reactions and reduce the cytotoxicity of these cells to achieve clinical efficacy[65] [Figure 1].
Figure 1.
This figure provides an insightful overview of various strategies that can be employed to effectively manage and deal with toxicity associated with CAR T cell therapy
Suicide strategies
To avoid unexpected side effects or eradicate transduced T cells in emergencies such as GVHD incidence or on-target off-tumor toxicity, the use of inducible safety switch genes could be an effective technique.[66] The selective and permanent elimination of CAR-T cells has been the subject of considerable investigation in recent years. Additionally, applying such strategies provides an effective solution to these challenges.[67]
HSV-TK
There has been some research exploring the use of Herpes simplex virus thymidine kinase (HSV-TK) in conjunction with CAR-T cell therapy. In this approach, the HSV-TK gene is inserted into the CAR-T cells themselves, allowing them to be targeted and eliminated if necessary, using ganciclovir (GCV).[68] This strategy can help address some of the safety concerns associated with CAR-T cell therapy, such as CRS or unexpected proliferation. The effectiveness and safety of using combination therapy to treat cancer are still being tested through experiments, so further research is necessary to determine its reliability.[69] The function of the suicide gene is to transform an inactive prodrug into an active and harmful one, which enables GCV to eradicate targeted cells.[70] HSV-TK phosphorylates the GCV and forms GCV-triphosphate, which binds to DNA as a triphosphate substrate. DNA replication is halted through GCV-induced chain termination. The HSV-TK/GCV compound can also trigger signaling cascades by forming Fas-associated death domain protein (FADD), leading to cellular death.[71]
EGFRt and CD20
A different type of safety mechanism for suicide involves modifying T cells to express surface antigens like truncated epidermal growth factor receptor (EGFRt) or CD20, which can be specifically targeted. As a result of this modification, the CAR-T cells are susceptible to either antibody-dependent cell-mediated cytotoxicity (ADCC) or complement-dependent cytotoxicity (CDC).[72] This susceptibility means that the CAR-T cells can be efficiently eliminated by administering monoclonal antibodies such as cetuximab (an EGFR-specific mAb) or rituximab (which is a CD20-specific mAb).[73]
iCasp9
The safety suicide gene known as inducible caspase 9 (iCasp9) is created by fusing human caspase 9 with FK506 binding protein (FKBP), a drug-sensitive protein.[74] When exposed to the chemical inducer of dimerization (AP1903), the fusion protein dimerizes, causing CAR-T cells to undergo apoptosis through the activation of downstream effector caspases.[75,76] The most recent variant of CAR-T cells has been engineered to focus on the CD20 antigen, and it incorporates both a truncated CD19 (ΔCD19) and an iCasp9 suicide gene. These altered T cells can eliminate tumor cells that exhibit CD20, and they could also be employed as a precautionary measure for patients diagnosed with B-cell-related cancers.[75] Administering iCasp9/AP1903 has been proven to effectively eliminate anti-CD20 CAR-T cells in a dose-dependent manner, making it a useful clinical approach.[77] Incorporating the IL-15 gene and an iCasp9-based suicide gene in T lymphocytes that display CAR targeting the CD19 antigen is a novel approach that can effectively enhance the anti-lymphoma/leukemia impact of CAR-T cells in individuals with B-cell-related cancers. This approach has been shown to be safe.[78]
Endogenous switch
iCARs
The inhibitory chimeric antigen receptors (iCARs) were created based on the concept of natural immune-inhibitory receptors found in T-cell regulation.[79] The iCARs have surface antigen recognition components and internal inhibitory signaling domains that come from different receptors such as PD1, CTLA-4, LAG-3, BTLA, and 2B4. These receptors help limit T cell responses even when an activating receptor is engaged.[79,80] Studies have shown that human T cells expressing iCARs can reduce cytokine secretion, cytotoxicity, and proliferation driven by endogenous TCRs or the CD19-specific receptor 19-28ζ.[81] In addition, an in vivo model was developed to demonstrate that iCAR-expressing T cells spare human cells expressing the iCAR antigen while still eliminating on-target, CD19-positive tumor cells but not off-target CD19 and iCAR antigen dual-positive cells.
Combinatorial targeting tumor-associated antigens
There is a significant desire to create “dual-targeted” T cells that have been altered to express two CARs: one with CD3ζ-linked scFv and another with a chimeric co-stimulatory receptor (CCR) containing CD28 and 4-1BB domains.[75] The co-transduced CAR-T cells could recognize two cancer-specific markers with separate signals and exhibit significant cytotoxicity, with minimal side effects on normal tissues.[82] The use of dual-CARs has advantages in combining different costimulatory domains. In leukemia experiments, using two CARs that target CD19 and CD22 with dual targeting showed better results than administering CD19 or CD22 monospecific CAR-T cells sequentially. A combination of a 28ζ and a BBζ CAR was more effective than having two 28ζ or two BBζ CARs. The best treatment outcomes occurred when the 28ζ CAR targeted CD19 and the BBζ CAR targeted CD22. Shalabi et al.[83] also demonstrated the superiority of combining 19.28ζ and 22.BBζ. Ruella et al.[13] used a bicistronicity vector encoding CD19 and CD123 BBζ CARs to show that the dual-CAR was superior to pooled CD19 and CD123 CAR-T cells.
In addition, Tan CAR-T cells are designed with two tandem linked scFvs that have specificity for different tumor-associated antigens simultaneously and coupled with the expressing serial CD28 and CD3ζ endo-domains.[84] Based on the strategy, combined target-antigen recognition ensures that CARs are activated only on tumor cells, thereby enhancing anti-tumor accuracy and reducing antigen escape.[85] Recent studies have found that targeting multiple antigens using modified CAR-T cells can be more effective in controlling heterogeneous tumors in vitro and in vivo.[86] One study found that a combination of CD19 and CD20 using the Tan-19.20 CAR was effective in controlling different types of tumors.[87] In another research, combining BCMA and GPRC5D CARs to target heterogeneous tumors showed that pooled BBζ and dual-BBζ CAR-T cells were more effective than Tan-BCMA.GPRC5D.BBζ counterparts. In preclinical models of glioblastoma, HER2.28ζ and IL13Rα2.28ζ Dual-CAR-T cells were superior to pooled monospecific CAR-T cells.[88] However, an IL13-mutein/HER2.scFv Tan-CAR allowed for more efficient synapse formation and better tumor control.[89] Combining CD19 and CD79a with Tan- or Dual-BBζ CARs was more effective than pooled monospecific CARs for preventing antigen escape, although there were some compromises in antigen binding and signaling.[90] Conversely, the Dual-CAR demonstrated reduced signaling, possibly due to competition for downstream signaling molecules.[89] Dual-CARs that use a common hinge/transmembrane component can heterodimerize and function as parallel CARs.
Synthetic notch receptors
Hacking endogenous T-cell response programs with synthetic Notch (synNotch) receptors provides new and more innovative cellular therapeutics to modulate the activity of the immune system toward cancer.[91] SynNotch circuits, as a programmable platform can be used to flexibly sculpt and customize cellular programs in naive T cells to extend the novel, non-native responses, overcoming tumor immunosuppression.[92] Synthetic Notch-based systems have been developed based on the wild-type Notch protein, consisting of an extracellular antigen recognition domain (e.g., single-chain antibodies or nanobodies), a transmembrane core regulatory domain (TMD), and a synthetic intracellular effector region (e.g., Gal4-VP64).[93] A recent structural study demonstrated that adding a hydrophobic natural motif (QHGQLWF) behind the C-terminus of the Notch core could significantly suppress ligand-independent activation (LIA) of synNotch receptors and refactor T cell functions.[94] Upon SynNotch T cells recognize the specific tumor antigen, conformational changes of the Notch core led to the proteolytic cleavage by the metalloproteinase, releasing the transcriptional regulator from the cytoplasmic tail and then triggering the expression of relevant target genes.[95] The redirected T cells can identify tumor antigens and effectively modify the surrounding microenvironments in a strong and well-regulated manner. The SynNotch receptors have the potential to guide the differentiation of T cells toward Th1 fate choice, leading to the production of IFNγ, which plays a crucial role in eradicating tumors.[91] Customized T cells can locally deliver diverse therapeutic payloads such as cytokines (IL-12 and MIP-1a), antibodies (α-PD-1, CTLA-4, BiTEs), and cytotoxic agents (TRAIL) therefore can minimize toxicity and increase therapeutic efficacy.[96]
CRISPR-associated CAR-T cells
The CRISPR/Cas9 system, which is the ultimate gene editor, has demonstrated remarkable efficacy and enormous potential in adoptive T-cell therapy.[97] This technique has many potential advantages, including high efficiency, flexibility, and multiplex genome editing, and opens a window to developing allogeneic universal CAR-T cells with more optimized structure and reduced toxicities.[98] Table 1 describes the clinical studies on this novel strategy.
Table 1.
Clinical Trials of CAR-T Cell Therapy with CRISPR-associated safety strategy
Clinical trial Identifier | Status | Target | Gene-knocked Out | Aim of study | Phase | Disease | ||||||
---|---|---|---|---|---|---|---|---|---|---|---|---|
NCT04037566 | Recruiting | CD19 | HPK1 | Safety | Phase 1 | Relapsed/Refractory Leukemia, B-Cell CD19 Positive | ||||||
NCT04637763 | Recruiting | CD19 | PD-1 | Safety, emerging efficacy, pharmacokinetics, & immunogenicity | Phase 1 | Relapsed/Refractory B Cell Non-Hodgkin Lymphoma | ||||||
NCT05722418 | Recruiting | BCMA | TRAC | Safety, effectiveness, determining the best dose | Phase 1 | Relapsed/Refractory Multiple Myeloma | ||||||
NCT05812326 | Completed | MUC1 | PD-1 | Safety, tolerability, & preliminary efficacy | Phase 1 Phase 2 |
Breast Cancer | ||||||
NCT04976218 | Recruiting | EGFR | TGFβR | Anti-tumor activities & safety | Phase 1 | EGFR-positive Solid Tumors | ||||||
NCT04767308 | Not yet recruiting | CD5 | CD5 | Safety & Efficacy | Early Phase 1 | Relapsed/Refractory CD5+Hematopoietic Malignancies | ||||||
NCT04502446 | Recruiting | CD70 | CD70 | Safety & Efficacy | Phase 1 | Relapsed/Refractory T or B Cell Malignancies | ||||||
NCT05397184 | Recruiting | CD3 and CD7 | TCR, CD7 & CD52 | Safety | Phase 1 | Relapsed/Refractory T-cell leukemia | ||||||
NCT04557436 | Recruiting | CD19 | CD52 & TRAC loci | Safety & Efficacy | Phase 1 | Relapsed/Refractory B Cell Acute Lymphoblastic Leukemia | ||||||
NCT05643742 | Recruiting | CD19 | CD19 | Safety & Efficacy | Phase 1 Phase 2 |
Relapsed/Refractory B-cell malignancies. | ||||||
NCT04438083 | Active, not recruiting | CD70 | CD83 | Safety & Efficacy | Phase 1 | Relapsed/Refractory renal cell carcinoma | ||||||
NCT05795595 | Recruiting | CD70 | CD70 | Safety & Efficacy | Phase 1 Phase 2 |
Relapsed/Refractory solid tumors |
Recent breakthroughs in allogeneic gene-edited CAR-T cells can circumvent the constraints and significant barriers such as GVHD and alloreactivity (host versus graft response).[99] CRISPR/Cas9 was used to successfully knock out the endogenous TRAC or TRBC locus (TCRα or TCRβ constant) and HLA-A or Beta-2-Microglobulin (B2M) locus, which was simultaneously replaced by incorporating the CAR cassette to generate double-knockout allogeneic universal anti-CD19 CAR T (DKO UCART19) cells to prevent TCR-induced GVHD, rapid rejection, and the risks of insertional oncogenesis.[15,100] Furthermore, it is advisable to incorporate HLA-E into the engineering of T cells to prevent rejection of CAR-T cells lacking HLA-I by Natural killer (NK) cells.[101]
A recent investigation demonstrated that CRISPR/Cas9-mediated knock-out of GM-CSF significantly reduced the risk of inflammation and CAR-T cell-induced severe CRS as well as enhanced anti-tumor activity.[102] Additionally, disrupting Diacylglycerol Kinase (DGK) in CAR-T cells targeting EGFRvIII could enhance resistance to soluble immunosuppressive factors and promote persistent antigen-specific cytotoxicity.[103] Various studies have demonstrated that the selective elimination of the CD7 gene, prominently expressed in T cells, through CRISPR/Cas9 editing significantly decreased the ability of CAR-T cells to kill other T cells in cases of T-cell acute lymphoblastic leukemia (T-ALL). This led to outstanding and long-lasting clinical responses, while also enhancing anti-cancer immunity and expanding the therapeutic range.[104]
Exogenous switch
Reversible ON-switch CARs
Intracellular micromolecule linkage is a remote-control strategy that addresses a general problem of excessive activity over-engineered therapeutic T cells. Researchers designed robust “ON-switch” CARs utilizing split synthetic receptors.[105] In this control system, the dual-input gated CAR construct resembles a logic gate that distributes key signaling modules from the conventional CAR into two separate components, extracellular single-chain antibody domain (scFv), and intracellular signaling domains. These split chimeric receptors only assemble in the presence of a heterodimerizing small molecule as an exogenous, co-activation signal.[106] The activation of CAR-T cells with molecule-controlled configuration could be controlled precisely by titratable pharmacologic regulation and then mitigating toxicity for durable CAR-T cell therapy.[107]
Juillerat and colleagues suggested the concept of “transient CAR-T cells,” which can be activated by the addition of a small molecule drug called AP21967 at specific locations and times. These modified CAR T-cells have multi-chain CARs that allow them to switch on or off the interaction between scFv/antigens and exhibit anti-tumor cytotoxicity.[108] The ability to regulate T cells is critical in enhancing the safety of this technology.[108]
Bispecific T cell-redirecting antibodies
BiTEs are created with the purpose of targeting a specific tumor-associated antigen (TAA) and CD3 co-receptor complexes using two separate scFvs that are linked together by flexible linker peptides.[109] These recombinant antibody-based bifunctional switches have high affinity and flexibility, along with many functional and pharmacological characteristics, which are responsible for CAR-T cell-engaging therapies’ long-term and improved efficacy.[110] Peptide-specific switchable CAR-T cells (sCAR-T) have been developed to recognize antigens through binding to the Ab-peptide neo-epitope (PNE) conjugate and the formation of immunological synapses, resulting in MHC-independent cancer cell elimination.[111]
Ab-fluorescein isothiocyanate (FITC) is a platform of bispecific T cell-redirecting antibodies that selectively regulate and recruit anti-FITC-CAR-T cells against tumor cells.[112] Various antibodies, in particular, trastuzumab, cetuximab, and rituximab could conjugate with FITC as supportive effectors and lead to targeted CAR-T cell-mediated cancer cell killing, along with dose-limiting cytokine release.[113] Blinatumomab, the first-in-class BiTE targeting CD19, has received full approval for the treatment of relapsed or refractory B-cell precursor acute lymphoblastic leukemia (R/R B-ALL) and appears to have curative potential in adult and pediatric patients with minimal residual disease (MRD) who still have detectable cancer traces after chemotherapy.[114] In addition to B-cell malignancies, BiTE therapy might be effective for a wide range of malignancies, including multiple solid cancers, and pave the way for the development of a more effective immunotherapeutic armamentarium.[115]
CONCLUSION
Genetically engineered CAR-T cell therapy has emerged as a leading option among adoptive cell treatments, particularly for hematopoietic malignancies.[43] The FDA’s approval of Tecartus (brexucabtagene autoleucel), a third-generation CAR-T therapy for mantle cell lymphoma (MCL), underscores its potential. However, the therapy is costly, complex to manufacture, and poses safety challenges, making it crucial to manage potential toxicities and identify biomarkers that predict side effects like CRS.[117] Continued analysis of overall survival data is necessary to accurately weigh the long-term benefits and risks associated with CAR-T cell therapy.
To enhance the efficacy of CAR-T therapy, researchers are focusing on identifying robust biomarkers in similar hematological malignancies.[118] Preclinical studies have shown promise with the B cell-activating factor receptor (BAFF-R), which exhibits cytotoxicity against various lymphoma and leukemia cell lines, including CD19-negative variants.[43] As the field progresses, there is a shift toward allogeneic CAR-T infusion due to the high costs and time commitment of traditional methods, with ongoing clinical trials exploring this avenue. Despite the significant advancements in CAR-T cell therapy, challenges remain in managing cytotoxicity and addressing scientific knowledge gaps that hinder widespread adoption.[119] The future will depend on improved medical management of adverse events and innovative gene therapy strategies to determine whether CAR-T therapy can become a first-line treatment for a broader range of hematologic malignancies and other neoplasms in the coming years.
Conflicts of interest
There are no conflicts of interest.
Funding Statement
Nil.
REFERENCES
- 1.Xu Z, Zeng S, Gong Z, Yan Y. Exosome-based immunotherapy: A promising approach for cancer treatment. Mol Cancer. 2020;19:1–16. doi: 10.1186/s12943-020-01278-3. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 2.Duan X, Chan C, Lin W. Nanoparticle-mediated immunogenic cell death enables and potentiates cancer immunotherapy. Angew Chem Int Ed Engl. 2019;58:670–80. doi: 10.1002/anie.201804882. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 3.Marofi F, Rahman HS, Thangavelu L, Dorofeev A, Bayas-Morejón F, Shirafkan N, et al. Renaissance of armored immune effector cells, CAR-NK cells, brings the higher hope for successful cancer therapy. Stem Cell Res Ther. 2021;12:1–21. doi: 10.1186/s13287-021-02251-7. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 4.Teoh PJ, Chng WJ. CAR T-cell therapy in multiple myeloma: More room for improvement. Blood Cancer J. 2021;11:1–18. doi: 10.1038/s41408-021-00469-5. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 5.Jin K-T, Chen B, Liu Y-Y, Yan J-P. Monoclonal antibodies and chimeric antigen receptor (CAR) T cells in the treatment of colorectal cancer. Cancer Cell Int. 2021;21:1–15. doi: 10.1186/s12935-021-01763-9. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 6.Boonserm P, Puthong S, Wichai T, Noitang S, Khunrae P, Sooksai S, et al. Investigation of major amino acid residues of anti-norfloxacin monoclonal antibodies responsible for binding with fluoroquinolones. Sci Rep. 2021;11:1–9. doi: 10.1038/s41598-021-96466-6. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 7.Benmebarek M-R, Karches CH, Cadilha BL, Lesch S, Endres S, Kobold S. Killing mechanisms of chimeric antigen receptor (CAR) T cells. Int J Mol Sci. 2019;20:1283. doi: 10.3390/ijms20061283. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 8.Rashidijahanabad Z, Huang X. Recent advances in tumor associated carbohydrate antigen based chimeric antigen receptor T cells and bispecific antibodies for anti-cancer immunotherapy. Semin Immunol. 2020;47:101390. doi: 10.1016/j.smim.2020.101390. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 9.Caruana I, Savoldo B, Hoyos V, Weber G, Liu H, Kim ES, et al. Heparanase promotes tumor infiltration and antitumor activity of CAR-redirected T lymphocytes. Nat Med. 2015;21:524–9. doi: 10.1038/nm.3833. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 10.Titov A, Petukhov A, Staliarova A, Motorin D, Bulatov E, Shuvalov O, et al. The biological basis and clinical symptoms of CAR-T therapy-associated toxicites. Cell Death Dis. 2018;9:1–15. doi: 10.1038/s41419-018-0918-x. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 11.Brentjens RJ, Davila ML, Riviere I, Park J, Wang X, Cowell LG, et al. CD19-targeted T cells rapidly induce molecular remissions in adults with chemotherapy-refractory acute lymphoblastic leukemia. Sci Transl Med. 2013;5:177ra38. doi: 10.1126/scitranslmed.3005930. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 12.Maus MV, June CH. Making better chimeric antigen receptors for adoptive T-cell therapy. Clin Cancer Res. 2016;22:1875–84. doi: 10.1158/1078-0432.CCR-15-1433. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 13.Ruella M, Barrett DM, Kenderian SS, Shestova O, Hofmann TJ, Perazzelli J, et al. Dual CD19 and CD123 targeting prevents antigen-loss relapses after CD19-directed immunotherapies. J Clin Invest. 2016;126:3814–26. doi: 10.1172/JCI87366. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 14.Zah E, Lin M-Y, Silva-Benedict A, Jensen MC, Chen YY. T cells expressing CD19/CD20 bispecific chimeric antigen receptors prevent antigen escape by malignant B cells bispecific T cells prevent B-cell antigen escape. Cancer Immunol Res. 2016;4:498–508. doi: 10.1158/2326-6066.CIR-15-0231. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 15.Eyquem J, Mansilla-Soto J, Giavridis T, van der Stegen SJ, Hamieh M, Cunanan KM, et al. Targeting a CAR to the TRAC locus with CRISPR/Cas9 enhances tumour rejection. Nature. 2017;543:113–7. doi: 10.1038/nature21405. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 16.Liu X, Zhang Y, Cheng C, Cheng AW, Zhang X, Li N, et al. CRISPR-Cas9-mediated multiplex gene editing in CAR-T cells. Cell Res. 2017;27:154–7. doi: 10.1038/cr.2016.142. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 17.Dimitri A, Herbst F, Fraietta JA. Engineering the next-generation of CAR T-cells with CRISPR-Cas9 gene editing. Mol Cancer. 2022;21:78. doi: 10.1186/s12943-022-01559-z. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 18.Mohanty R, Chowdhury CR, Arega S, Sen P, Ganguly P, Ganguly N. CAR T cell therapy: A new era for cancer treatment (Review) Oncol Rep. 2019;42:2183–95. doi: 10.3892/or.2019.7335. [DOI] [PubMed] [Google Scholar]
- 19.Mardomi A, Abediankenari SJCM. Matrix metalloproteinase 8: Could it benefit the CAR-T cell therapy of solid tumors?-A-Commentary on therapeutic potential. Cancer Microenviron. 2018;11:93–6. doi: 10.1007/s12307-018-0208-2. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 20.Wang Z, Wu Z, Liu Y, Han W. New development in CAR-T cell therapy. J Hematol Oncol. 2017;10:1–11. doi: 10.1186/s13045-017-0423-1. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 21.Harrison RA, Tummala S, de Groot J. Neurologic toxicities of cancer immunotherapies: A review. Curr Neurol Neurosci Rep. 2020;20:1–11. doi: 10.1007/s11910-020-01038-2. [DOI] [PubMed] [Google Scholar]
- 22.Azam U, Sridharan K. CAR-T cell clinical trials experience–Past, present and future. Second Generation Cell and Gene-Based Therapies. 2020:303–75. [Google Scholar]
- 23.Hernández-López A, Téllez-González MA, Mondragón-Terán P, Meneses-Acosta A. Chimeric antigen receptor-T cells: A pharmaceutical scope. Front Pharmacol. 2021;12:720692. doi: 10.3389/fphar.2021.720692. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 24.Cosenza M, Sacchi S, Pozzi S. Cytokine release syndrome associated with T-cell-based therapies for hematological malignancies: Pathophysiology, clinical presentation, and treatment. Int J Mol Sci. 2021;22:7652. doi: 10.3390/ijms22147652. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 25.Lee DW, Gardner R, Porter DL, Louis CU, Ahmed N, Jensen M, et al. Current concepts in the diagnosis and management of cytokine release syndrome. Blood. 2014;124:188–95. doi: 10.1182/blood-2014-05-552729. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 26.Grupp SA, Porter DL, Teachey DT, Barrett DM, Chew A, Suppa E, et al. CD19-redirected chimeric antigen receptor T (CART19) cells induce a cytokine release syndrome (CRS) and induction of treatable macrophage activation syndrome (MAS) that can be managed by the IL-6 antagonist tocilizumab (toc) Blood. 2012;120:2604. [Google Scholar]
- 27.Iqubal A, Hoda F, Najmi AK, Haque SE. Macrophage activation and cytokine release syndrome in COVID-19: Current updates and analysis of repurposed and investigational anti-cytokine drugs. Drug Res (Stuttg) 2021;71:173–9. doi: 10.1055/a-1291-7692. [DOI] [PubMed] [Google Scholar]
- 28.Maude SL, Barrett D, Teachey DT, Grupp SA. Managing cytokine release syndrome associated with novel T cell-engaging therapies. Cancer J (Sudbury, Mass) 2014;20:119–22. doi: 10.1097/PPO.0000000000000035. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 29.Lee DW, Kochenderfer JN, Stetler-Stevenson M, Cui YK, Delbrook C, Feldman SA, et al. T cells expressing CD19 chimeric antigen receptors for acute lymphoblastic leukaemia in children and young adults: A phase 1 dose-escalation trial. Lancet. 2015;385:517–28. doi: 10.1016/S0140-6736(14)61403-3. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 30.Davila ML, Riviere I, Wang X, Bartido S, Park J, Curran K, et al. Efficacy and toxicity management of 19-28z CAR T cell therapy in B cell acute lymphoblastic leukemia. Sci Transl Med. 2014;6:224ra25. doi: 10.1126/scitranslmed.3008226. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 31.Maude SL, Frey N, Shaw PA, Aplenc R, Barrett DM, Bunin NJ, et al. Chimeric antigen receptor T cells for sustained remissions in leukemia. N Engl J Med. 2014;371:1507–17. doi: 10.1056/NEJMoa1407222. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 32.Kotch C, Barrett D, Teachey DT. Tocilizumab for the treatment of chimeric antigen receptor T cell-induced cytokine release syndrome. Expert Rev Clin Immunol. 2019;15:813–22. doi: 10.1080/1744666X.2019.1629904. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 33.Freyer CW, Porter DL. Cytokine release syndrome and neurotoxicity following CAR T-cell therapy for hematologic malignancies. J Allergy Clin Immunol. 2020;146:940–8. doi: 10.1016/j.jaci.2020.07.025. [DOI] [PubMed] [Google Scholar]
- 34.Thakar MS, Kearl TJ, Malarkannan S. Controlling cytokine release syndrome to harness the full potential of CAR-based cellular therapy. Front Oncol. 2019;9:1529. doi: 10.3389/fonc.2019.01529. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 35.Kochenderfer JN, Dudley ME, Feldman SA, Wilson WH, Spaner DE, Maric I, et al. B-cell depletion and remissions of malignancy along with cytokine-associated toxicity in a clinical trial of anti-CD19 chimeric-antigen-receptor–transduced T cells. Blood. 2012;119:2709–20. doi: 10.1182/blood-2011-10-384388. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 36.Porter DL, Levine BL, Kalos M, Bagg A, June CH. Chimeric antigen receptor–modified T cells in chronic lymphoid leukemia; Chimeric antigen receptor-modified T cells for acute lymphoid leukemia; Chimeric antigen receptor T cells for sustained remissions in leukemia. N Engl J Med. 2011;365:725–33. doi: 10.1056/NEJMoa1103849. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 37.Miao L, Zhang Z, Ren Z, Li Y. Reactions related to CAR-T cell therapy. Front Immunol. 2021;12:663201. doi: 10.3389/fimmu.2021.663201. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 38.Patel C, Wilson CP, Ahmed N, Hattab Y. Acute uric acid nephropathy following epileptic seizures: Case report and review. Case Rep Nephrol. 2019;2019:4890287. doi: 10.1155/2019/4890287. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 39.Khammissa RAG, Fourie J, Motswaledi MH, Ballyram R, Lemmer J, Feller L. The biological activities of vitamin D and its receptor in relation to calcium and bone homeostasis, cancer, immune and cardiovascular systems, skin biology, and oral health. Biomed Res Int. 2018;2018:9276380. doi: 10.1155/2018/9276380. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 40.Yáñez L, Sánchez-Escamilla M, Perales M-A. CAR T cell toxicity: Current management and future directions. Hemasphere. 2019;3:e186. doi: 10.1097/HS9.0000000000000186. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 41.Acharya UH, Dhawale T, Yun S, Jacobson CA, Chavez JC, Ramos JD, et al. Management of cytokine release syndrome and neurotoxicity in chimeric antigen receptor (CAR) T cell therapy. Expert Rev Hematol. 2019;12:195–205. doi: 10.1080/17474086.2019.1585238. [DOI] [PubMed] [Google Scholar]
- 42.Belay Y, Yirdaw K, Enawgaw B. Tumor lysis syndrome in patients with hematological malignancies. J Oncol. 2017;2017:9684909. doi: 10.1155/2017/9684909. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 43.Han D, Xu Z, Zhuang Y, Ye Z, Qian Q. Current progress in CAR-T cell therapy for hematological malignancies. J Cancer. 2021;12:326–34. doi: 10.7150/jca.48976. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 44.Marofi F, Motavalli R, Safonov VA, Thangavelu L, Yumashev AV, Alexander M, et al. CAR T cells in solid tumors: Challenges and opportunities. Stem Cell Res Ther. 2021;12:1–16. doi: 10.1186/s13287-020-02128-1. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 45.Blanc V, Bousseau A, Caron A, Carrez C, Lutz RJ, Lambert JM. SAR3419: An anti-CD19-maytansinoid immunoconjugate for the treatment of B-cell malignancies. Clin Cancer Res. 2011;17:6448–58. doi: 10.1158/1078-0432.CCR-11-0485. [DOI] [PubMed] [Google Scholar]
- 46.Salter AI, Pont MJ, Riddell SR. Chimeric antigen receptor–modified T cells: CD19 and the road beyond. Blood. 2018;131:2621–9. doi: 10.1182/blood-2018-01-785840. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 47.Leandro MJ, Edwards JC, Cambridge G. Clinical outcome in 22 patients with rheumatoid arthritis treated with B lymphocyte depletion. Ann Rheum Dis. 2002;61:883–8. doi: 10.1136/ard.61.10.883. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 48.Gonultas F, Akbulut S, Barut B, Kutluturk K, Yilmaz S. Graft-versus-host disease after living donor liver transplantation: An unpredictable troublesome complication for liver transplant centers. Eur J Gastroenterol Hepatol. 2020;32:95–100. doi: 10.1097/MEG.0000000000001530. [DOI] [PubMed] [Google Scholar]
- 49.Duarte LRF, Pinho V, Rezende BM, Teixeira MM. Resolution of inflammation in acute graft-versus-host-disease: Advances and perspectives. Biomolecules. 2022;12:75. doi: 10.3390/biom12010075. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 50.Wu Y, Huang Z, Harrison R, Liu L, Zhu L, Situ Y, et al. Engineering CAR T cells for enhanced efficacy and safety. APL Bioeng. 2022;6:011502. doi: 10.1063/5.0073746. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 51.Huan T, Chen D, Liu G, Zhang H, Wang X, Wu Z, et al. Activation-induced cell death in CAR-T cell therapy. Hum Cell. 2022;35:441–7. doi: 10.1007/s13577-022-00670-z. [DOI] [PubMed] [Google Scholar]
- 52.Ventosa Nivet I. CD19-targeted CAR T cells Therapy. 2020 [Google Scholar]
- 53.Jellusova J. The role of metabolic checkpoint regulators in B cell survival and transformation. Immunol Rev. 2020;295:39–53. doi: 10.1111/imr.12855. [DOI] [PubMed] [Google Scholar]
- 54.Shum T, Kruse RL, Rooney CM. Strategies for enhancing adoptive T-cell immunotherapy against solid tumors using engineered cytokine signaling and other modalities. Expert Opin Biol Ther. 2018;18:653–64. doi: 10.1080/14712598.2018.1473368. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 55.Miliotou AN, Papadopoulou LC. CAR T-cell therapy: A new era in cancer immunotherapy. Curr Pharm Biotechnol. 2018;19:5–18. doi: 10.2174/1389201019666180418095526. [DOI] [PubMed] [Google Scholar]
- 56.Blind JE, McLeod EN, Campbell KJ. Viral-mediated gene therapy and genetically modified therapeutics: A primer on biosafety handling for the health-system pharmacist. Am J Health Syst Pharm. 2019;76:795–802. doi: 10.1093/ajhp/zxz056. [DOI] [PubMed] [Google Scholar]
- 57.Hacein-Bey-Abina S, Garrigue A, Wang GP, Soulier J, Lim A, Morillon E, et al. Insertional oncogenesis in 4 patients after retrovirus-mediated gene therapy of SCID-X1. J Clin Invest. 2008;118:3132–42. doi: 10.1172/JCI35700. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 58.Stock S, Schmitt M, Sellner L. Optimizing manufacturing protocols of chimeric antigen receptor T cells for improved anticancer immunotherapy. Int J Mol Sci. 2019;20:6223. doi: 10.3390/ijms20246223. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 59.Scholler J, Brady TL, Binder-Scholl G, Hwang W-T, Plesa G, Hege KM, et al. Decade-long safety and function of retroviral-modified chimeric antigen receptor T cells. Sci Transl Med. 2012;4:132ra53. doi: 10.1126/scitranslmed.3003761. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 60.Raes L, De Smedt SC, Raemdonck K, Braeckmans K. Non-viral transfection technologies for next-generation therapeutic T cell engineering. Biotechnol Adv. 2021;49:107760. doi: 10.1016/j.biotechadv.2021.107760. [DOI] [PubMed] [Google Scholar]
- 61.Siegler EL, Kenderian SS. Neurotoxicity and cytokine release syndrome after chimeric antigen receptor T cell therapy: Insights into mechanisms and novel therapies. Front Immunol. 2020;11:1973. doi: 10.3389/fimmu.2020.01973. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 62.Rice J, Nagle S, Randall J, Hinson HE. Chimeric antigen receptor T cell-related neurotoxicity: Mechanisms, clinical presentation, and approach to treatment. Curr Treat Options Neurol. 2019;21:40. doi: 10.1007/s11940-019-0580-3. [DOI] [PubMed] [Google Scholar]
- 63.Gust J, Finney OC, Li D, Brakke HM, Hicks RM, Futrell RB, et al. Glial injury in neurotoxicity after pediatric CD19-directed chimeric antigen receptor T cell therapy. Ann Neurol. 2019;86:42–54. doi: 10.1002/ana.25502. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 64.Neelapu SS. Managing the toxicities of CAR T-cell therapy. Hematol Oncol. 2019;37(Suppl 1):48–52. doi: 10.1002/hon.2595. [DOI] [PubMed] [Google Scholar]
- 65.Fischer JW, Bhattarai N. CAR-T cell therapy: Mechanism, management, and mitigation of inflammatory toxicities. Front Immunol. 2021;12:693016. doi: 10.3389/fimmu.2021.693016. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 66.Casucci M, Falcone L, Camisa B, Norelli M, Porcellini S, Stornaiuolo A, et al. Extracellular NGFR spacers allow efficient tracking and enrichment of fully functional CAR-T cells co-expressing a suicide gene. Front Immunol. 2018;9:507. doi: 10.3389/fimmu.2018.00507. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 67.Safarzadeh Kozani P, Safarzadeh Kozani P, Rahbarizadeh F, Khoshtinat Nikkhoi S. Strategies for dodging the obstacles in CAR T cell therapy. Front Oncol. 2021;11:627549. doi: 10.3389/fonc.2021.627549. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 68.Ingemarsdotter CK, Poddar S, Mercier S, Patzel V, Lever AML. Expression of herpes simplex virus thymidine kinase/ganciclovir by RNA trans-splicing induces selective killing of HIV-producing cells. Mol Ther Nucleic Acids. 2017;7:140–54. doi: 10.1016/j.omtn.2017.03.004. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 69.Greco R, Oliveira G, Stanghellini MTL, Vago L, Bondanza A, Peccatori J, et al. Improving the safety of cell therapy with the TK-suicide gene. Front Pharmacol. 2015;6:95. doi: 10.3389/fphar.2015.00095. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 70.Moolten FL, Wells JM. Curability of tumors bearing herpes thymidine kinase genes transfered by retroviral vectors. J Natl Cancer Inst. 1990;82:297–300. doi: 10.1093/jnci/82.4.297. [DOI] [PubMed] [Google Scholar]
- 71.Sangro B, Mazzolini G, Ruiz M, Ruiz J, Quiroga J, Herrero I, et al. A phase I clinical trial of thymidine kinase-based gene therapy in advanced hepatocellular carcinoma. Cancer Gene Ther. 2010;17:837–43. doi: 10.1038/cgt.2010.40. [DOI] [PubMed] [Google Scholar]
- 72.Paszkiewicz PJ, Fräßle SP, Srivastava S, Sommermeyer D, Hudecek M, Drexler I, et al. Targeted antibody-mediated depletion of murine CD19 CAR T cells permanently reverses B cell aplasia. J Clin Invest. 2016;126:4262–72. doi: 10.1172/JCI84813. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 73.Griffioen M, van Egmond EH, Kester MG, Willemze R, Falkenburg JF, Heemskerk MH. Retroviral transfer of human CD20 as a suicide gene for adoptive T-cell therapy. Haematologica. 2009;94:1316–20. doi: 10.3324/haematol.2008.001677. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 74.Mirzaee Godarzee M, Mahmud Hussen B, Razmara E, Hakak-Zargar B, Mohajerani F, Dabiri H, et al. Strategies to overcome the side effects of chimeric antigen receptor T cell therapy. Ann N Y Acad Sci. 2022;1510:18–35. doi: 10.1111/nyas.14724. [DOI] [PubMed] [Google Scholar]
- 75.Di Stasi A, Tey S-K, Dotti G, Fujita Y, Kennedy-Nasser A, Martinez C, et al. Inducible apoptosis as a safety switch for adoptive cell therapy. N Engl J Med. 2011;365:1673–83. doi: 10.1056/NEJMoa1106152. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 76.Gargett T, Brown MP. The inducible caspase-9 suicide gene system as a “safety switch” to limit on-target, off-tumor toxicities of chimeric antigen receptor T cells. Front Pharmacol. 2014;5:235. doi: 10.3389/fphar.2014.00235. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 77.Budde LE, Berger C, Lin Y, Wang J, Lin X, Frayo SE, et al. Combining a CD20 chimeric antigen receptor and an inducible caspase 9 suicide switch to improve the efficacy and safety of T cell adoptive immunotherapy for lymphoma. PLoS One. 2013;8:e82742. doi: 10.1371/journal.pone.0082742. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 78.Hoyos V, Savoldo B, Quintarelli C, Mahendravada A, Zhang M, Vera J, et al. Engineering CD19-specific T lymphocytes with interleukin-15 and a suicide gene to enhance their anti-lymphoma/leukemia effects and safety. Leukemia. 2010;24:1160–70. doi: 10.1038/leu.2010.75. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 79.Rafiq S, Yeku OO, Jackson HJ, Purdon TJ, Van Leeuwen DG, Drakes DJ, et al. Targeted delivery of a PD-1-blocking scFv by CAR-T cells enhances anti-tumor efficacy in vivo. Nat Biotechnol. 2018;36:847–56. doi: 10.1038/nbt.4195. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 80.Liu H, Lei W, Zhang C, Yang C, Wei J, Guo Q, et al. CD19-specific CAR T cells that express a PD-1/CD28 chimeric switch-receptor are effective in patients with PD-L1-positive B-cell lymphoma. Clin Cancer Res. 2021;27:473–84. doi: 10.1158/1078-0432.CCR-20-1457. [DOI] [PubMed] [Google Scholar]
- 81.Luo Y, Song G, Liang S, Li F, Liu K. Research advances in chimeric antigen receptor-modified T-cell therapy (Review) Exp Ther Med. 2021;21:484. doi: 10.3892/etm.2021.9915. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 82.Kloss CC, Condomines M, Cartellieri M, Bachmann M, Sadelain M. Combinatorial antigen recognition with balanced signaling promotes selective tumor eradication by engineered T cells. Nat Biotechnol. 2013;31:71–5. doi: 10.1038/nbt.2459. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 83.Shalabi H, Qin H, Su A, Yates B, Wolters PL, Steinberg SM, et al. CD19/22 CAR T cells in children and young adults with B-ALL: Phase 1 results and development of a novel bicistronic CAR. Blood. 2022;140:451–63. doi: 10.1182/blood.2022015795. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 84.Grada Z, Hegde M, Byrd T, Shaffer DR, Ghazi A, Brawley VS, et al. TanCAR: A novel bispecific chimeric antigen receptor for cancer immunotherapy. Mol Ther Nucleic Acids. 2013;2:e105. doi: 10.1038/mtna.2013.32. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 85.Schneider D, Xiong Y, Wu D, Nölle V, Schmitz S, Haso W, et al. A tandem CD19/CD20 CAR lentiviral vector drives on-target and off-target antigen modulation in leukemia cell lines. J Immunother Cancer. 2017;5:1–17. doi: 10.1186/s40425-017-0246-1. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 86.Shertzer H. Genetic Relationships and Therapeutic Options for Relapsed Acute Lymphoblastic Leukemia. 2020 [Google Scholar]
- 87.Fernández de Larrea C, Staehr M, Lopez AV, Ng KY, Chen Y, Godfrey WD, et al. Defining an optimal dual-targeted CAR T-cell therapy approach simultaneously targeting BCMA and GPRC5D to prevent BCMA escape–Driven relapse in multiple myeloma. Blood Cancer Discov. 2020;1:146–54. doi: 10.1158/2643-3230.BCD-20-0020. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 88.Schmidts A, Srivastava AA, Ramapriyan R, Bailey SR, Bouffard AA, Cahill DP, et al. Tandem chimeric antigen receptor (CAR) T cells targeting EGFRvIII and IL-13Rα2 are effective against heterogeneous glioblastoma. Neurooncol Adv. 2023;5:vdac185. doi: 10.1093/noajnl/vdac185. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 89.Leung I, Templeton ML, Lo Y, Rajan A, Stull SM, Garrison SM, et al. Compromised antigen binding and signaling interfere with bispecific CD19 and CD79a chimeric antigen receptor function. Blood Adv. 2022;7:2718–30. doi: 10.1182/bloodadvances.2022008559. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 90.Hudecek M, Sommermeyer D, Kosasih PL, Silva-Benedict A, Liu L, Rader C, et al. The nonsignaling extracellular spacer domain of chimeric antigen receptors is decisive for in vivo antitumor activity. Cancer Immunol Res. 2015;3:125–35. doi: 10.1158/2326-6066.CIR-14-0127. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 91.Wang Z, Wang F, Zhong J, Zhu T, Zheng Y, Zhao T, et al. Using apelin-based synthetic Notch receptors to detect angiogenesis and treat solid tumors. Nat Commun. 2020;11:2163. doi: 10.1038/s41467-020-15729-4. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 92.Roybal KT, Rupp LJ, Morsut L, Walker WJ, McNally KA, Park JS, et al. Precision tumor recognition by T cells with combinatorial antigen-sensing circuits. Cell. 2016;164:770–9. doi: 10.1016/j.cell.2016.01.011. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 93.Morsut L, Roybal KT, Xiong X, Gordley RM, Coyle SM, Thomson M, et al. Engineering customized cell sensing and response behaviors using synthetic notch receptors. Cell. 2016;164:780–91. doi: 10.1016/j.cell.2016.01.012. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 94.Yang Z-J, Yu Z-Y, Cai Y-M, Du R-R, Cai L. Engineering of an enhanced synthetic Notch receptor by reducing ligand-independent activation. Commun Biol. 2020;3:1–7. doi: 10.1038/s42003-020-0848-x. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 95.Choe JH, Watchmaker PB, Simic MS, Gilbert RD, Li AW, Krasnow NA, et al. SynNotch-CAR T cells overcome challenges of specificity, heterogeneity, and persistence in treating glioblastoma. Sci Transl Med. 2021;13:eabe7378. doi: 10.1126/scitranslmed.abe7378. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 96.Roybal KT, Williams JZ, Morsut L, Rupp LJ, Kolinko I, Choe JH, et al. Engineering T cells with customized therapeutic response programs using synthetic notch receptors. Cell. 2016;167:419–32. doi: 10.1016/j.cell.2016.09.011. e16. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 97.Ureña-Bailén G, Lamsfus-Calle A, Daniel-Moreno A, Raju J, Schlegel P, Seitz C, et al. CRISPR/Cas9 technology: Towards a new generation of improved CAR-T cells for anticancer therapies. Brief Funct Genomics. 2020;19:191–200. doi: 10.1093/bfgp/elz039. [DOI] [PubMed] [Google Scholar]
- 98.Li C, Mei H, Hu Y. Applications and explorations of CRISPR/Cas9 in CAR T-cell therapy. Brief Funct Genomics. 2020;19:175–82. doi: 10.1093/bfgp/elz042. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 99.Ou X, Ma Q, Yin W, Ma X, He Z. CRISPR/Cas9 gene-editing in cancer immunotherapy: Promoting the present revolution in cancer therapy and exploring more. Front Cell Dev Biol. 2021;9:674467. doi: 10.3389/fcell.2021.674467. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 100.Torikai H, Reik A, Soldner F, Warren EH, Yuen C, Zhou Y, et al. Toward eliminating HLA class I expression to generate universal cells from allogeneic donors. Blood. 2013;122:1341–9. doi: 10.1182/blood-2013-03-478255. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 101.Gornalusse GG, Hirata RK, Funk SE, Riolobos L, Lopes VS, Manske G, et al. HLA-E-expressing pluripotent stem cells escape allogeneic responses and lysis by NK cells. Nat Biotechnol. 2017;35:765–72. doi: 10.1038/nbt.3860. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 102.Sterner RM, Sakemura R, Cox MJ, Yang N, Khadka RH, Forsman CL, et al. GM-CSF inhibition reduces cytokine release syndrome and neuroinflammation but enhances CAR-T cell function in xenografts. Blood. 2019;133:697–709. doi: 10.1182/blood-2018-10-881722. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 103.Jung I-Y, Kim Y-Y, Yu H-S, Lee M, Kim S, Lee J. CRISPR/Cas9-mediated knockout of DGK improves antitumor activities of human T cells. Cancer Res. 2018;78:4692–703. doi: 10.1158/0008-5472.CAN-18-0030. [DOI] [PubMed] [Google Scholar]
- 104.Cooper ML, Choi J, Staser K, Ritchey JK, Devenport JM, Eckardt K, et al. An “off-the-shelf” fratricide-resistant CAR-T for the treatment of T cell hematologic malignancies. Leukemia. 2018;32:1970–83. doi: 10.1038/s41375-018-0065-5. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 105.Halim L, Ajina A, Maher J. Pre-clinical development of chimeric antigen receptor T-cell immunotherapy: Implications of design for efficacy and safety. Best Pract Res Clin Haematol. 2018;31:117–25. doi: 10.1016/j.beha.2018.04.002. [DOI] [PubMed] [Google Scholar]
- 106.Wu C-Y, Roybal KT, Puchner EM, Onuffer J, Lim WA. Remote control of therapeutic T cells through a small molecule–gated chimeric receptor. Science. 2015;350:aab4077. doi: 10.1126/science.aab4077. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 107.Jan M, Scarfò I, Larson RC, Walker A, Schmidts A, Guirguis AA, et al. Reversible ON- and OFF-switch chimeric antigen receptors controlled by lenalidomide. Sci Transl Med. 2021;13:eabb6295. doi: 10.1126/scitranslmed.abb6295. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 108.Juillerat A, Marechal A, Filhol J-M, Valton J, Duclert A, Poirot L, et al. Design of chimeric antigen receptors with integrated controllable transient functions. Sci Rep. 2016;6:18950. doi: 10.1038/srep18950. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 109.Verkleij CPM, Frerichs KA, Broekmans M, Absalah S, Maas-Bosman PWC, Kruyswijk S, et al. T-cell redirecting bispecific antibodies targeting BCMA for the treatment of multiple myeloma. Oncotarget. 2020;11:4076–81. doi: 10.18632/oncotarget.27792. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 110.Goebeler M-E, Bargou RC. T cell-engaging therapies—BiTEs and beyond. Nat Rev Clin Oncol. 2020;17:418–34. doi: 10.1038/s41571-020-0347-5. [DOI] [PubMed] [Google Scholar]
- 111.Rodgers DT, Mazagova M, Hampton EN, Cao Y, Ramadoss NS, Hardy IR, et al. Switch-mediated activation and retargeting of CAR-T cells for B-cell malignancies. Proc Natl Acad Sci U S A. 2016;113:E459–68. doi: 10.1073/pnas.1524155113. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 112.Kim MS, Ma JS, Yun H, Cao Y, Kim JY, Chi V, et al. Redirection of genetically engineered CAR-T cells using bifunctional small molecules. J Am Chem Soc. 2015;137:2832–5. doi: 10.1021/jacs.5b00106. [DOI] [PubMed] [Google Scholar]
- 113.Tamada K, Geng D, Sakoda Y, Bansal N, Srivastava R, Li Z, et al. Redirecting gene-modified T cells toward various cancer types using tagged antibodies. Clin Cancer Res. 2012;18:6436–45. doi: 10.1158/1078-0432.CCR-12-1449. [DOI] [PubMed] [Google Scholar]
- 114.Zhu M, Wu B, Brandl C, Johnson J, Wolf A, Chow A, et al. Blinatumomab, a bispecific T-cell engager (BiTE®) for CD-19 targeted cancer immunotherapy: Clinical pharmacology and its implications. Clin Pharmacokinet. 2016;55:1271–88. doi: 10.1007/s40262-016-0405-4. [DOI] [PubMed] [Google Scholar]
- 115.Scott EM, Duffy MR, Freedman JD, Fisher KD, Seymour LW. Solid tumor immunotherapy with T cell engager-armed oncolytic viruses. Macromol Biosci. 2018;18:1700187. doi: 10.1002/mabi.201700187. [DOI] [PubMed] [Google Scholar]
- 116.Zhao Q, Yu J, Meng X. A good start of immunotherapy in esophageal cancer. Cancer Med. 2019;8:4519–26. doi: 10.1002/cam4.2336. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 117.Mian A, Hill BT. Brexucabtagene autoleucel for the treatment of relapsed/refractory mantle cell lymphoma. Expert Opin Biol Ther. 2021;21:435–41. doi: 10.1080/14712598.2021.1889510. [DOI] [PubMed] [Google Scholar]
- 118.El-Khazragy N, Ghozy S, Emad P, Mourad M, Razza D, Farouk YK, et al. Chimeric antigen receptor T cells immunotherapy: Challenges and opportunities in hematological malignancies. Immunotherapy. 2020;12:1341–57. doi: 10.2217/imt-2020-0181. [DOI] [PubMed] [Google Scholar]
- 119.Qin H, Dong Z, Wang X, Cheng WA, Wen F, Xue W, et al. CAR T cells targeting BAFF-R can overcome CD19 antigen loss in B cell malignancies. Sci Transl Med. 2019;11:eaaw9414. doi: 10.1126/scitranslmed.aaw9414. [DOI] [PMC free article] [PubMed] [Google Scholar]