Abstract
Chimeric antigen receptor-natural killer (CAR-NK) cells have emerged as another prominent player in the realm of tumor immunotherapy following CAR-T cells. The unique features of CAR-NK cells make it possible to compensate for deficiencies in CAR-T therapy, such as the complexity of the manufacturing process, clinical adverse events, and solid tumor challenges. To date, CAR-NK products from different allogeneic sources have exhibited remarkable anti-tumor effects on preclinical studies and have gradually been applied in clinical practice. However, each source has advantages and disadvantages. Selecting a suitable source may help maximize CAR-NK cell efficacy and increase the feasibility of clinical transformation. Therefore, this review discusses the development and challenges of CAR-NK cells from different sources to provide a reference for future exploration.
Keywords: CAR-NK cells, cord blood, iPSC, NK-92 cells, peripheral blood
Introduction
Despite the remarkable effectiveness of chimeric antigen receptor-T (CAR-T) cell therapy in human malignancies, it is associated with a plethora of challenges (1). In addition to optimizing CAR-T cell efficacy, the alternative role of natural killer (NK) cells has garnered significant interest. The distinct characteristics of chimeric antigen receptor-natural killer (CAR-NK) cells highlight their superior potential for anti-tumor efficacy compared to CAR-T cells (2). In addition to CAR-mediated cytotoxicity, NK cells can lyse cancer cells with downregulated or deficient target antigens via NK-specific cytotoxicity, indicating that CAR-NK cells are excellent candidates for eliminating heterogeneous solid tumors (3). CAR-NK therapy is also believed to be safer than CAR-T cell therapy, given the short-term survival of CAR-NK cells in vivo and the distinctive cytokine spectrum compared to CAR-T cells (4). “Multiple sources” is another significant advantage of CAR-NK therapy. In addition to autologous peripheral blood-derived NK cells, multiple allogeneic NK cells further enhance the possibility of fabricating off-the-shelf CAR-NK products to help reduce patient waiting times and treatment costs (5). However, each source has its drawbacks. Therefore, in this review, different CAR-NK cell sources are discussed to assist in making an informed choice when applying CAR-NK cells.
Comparison of CAR-NK cells derived from different sources
Peripheral blood (PB), cord blood (CB), NK cell lines, and iPSC are the main sources of NK cells for CAR modification (5). NK cells from four promising cell types possess their advantages but also exhibit different problems upon application (Figure 1).
Figure 1.
Different sources for manufacturing CAR-NK cells. (A) PB-derived CAR-NK cells; (B) CB-derived CAR-NK cells; (C) NK cell line (NK-92)-derived CAR-NK cells; (D) iPSC-derived CAR-NK cells. A sufficient number of CAR-NK cells derived from the four allogeneic sources can be delivered to cancer through systemic or regional administration (e.g., pleural, peritoneal, and direct intra-tumoral injection). CAR-NK, chimeric antigen receptor-natural killer; PB, peripheral blood; PBMC, peripheral blood mononuclear cells; CB, cord blood; CBMC, cord blood mononuclear cells; aAPC, artificial antigen-presenting cells. The figure is created with BioRender.com.
PB-derived CAR-NK cells
PB-NK cells
Human PB mononuclear cells (PBMC) are conventionally used as NK cell sources in the early adoptive immunotherapy (6,7). PB-NK cells have two subsets (8): CD56brightCD16−/dim PB-NK cells that are the mostly immature and CD56dimCD16+ that are more mature. CD56brightCD16−/dim PB-NK has longer telomeres (9) and will gradually acquire CD56dim phenotypes (obtaining CD16 and KIRs and losing NKG2A) as it matures (10). PB-NK cells used for immunotherapy can be divided into autologous and allogeneic sources. Autologous NK cells usually exhibit more limited anti-tumor effects than allogeneic NK cells (11), since autologous KIR/HLA matching between NK and tumor cells and immunosuppressive conditions are critical contributors to NK cell inactivation and dysfunction (12). In contrast, mismatched KIR/HLA between allogeneic NK and tumor cells excludes interference of inhibitory signals and ensures sufficient NK activity (13). However, it should be noted that T cells in allogeneic peripheral blood must be removed before NK cell application to avoid graft versus host disease (GVHD) (14). As a popular choice for gene modification, universal accessibility, low cost, and high safety are the main advantages of PB-NK. The overexpression of killer cell-activating receptors in PB-NK, such as CD16, NKP44, and NKP46, also plays an essential role in enhancing anti-tumor activity (15).
Advances in research on PB-CAR-NK cells
PB-CAR-NK cells have demonstrated significant efficacy in treating both hematological and solid malignancies. The 2G CAR structure is the most popular. Kruschinski et al. constructed human epidermal growth factor receptor 2 (HER2)-specific CAR-NK cells with CD28-CD3ζ structure. This could override inhibitory signals in primary NK cells and exhibit high-level cytokine release and degranulation to eradicate tumor cells in HER2-positive carcinoma models (16). Similarly, Imai et al. demonstrated that anti-CD19 CAR-NK cells with 4-1BB-CD3ζ structure were capable of surmounting NK resistance and producing incremental amounts of INF-γ and GM-CSF to promote the killing of leukemic cells (17). Recently, additional NK cell-specific CAR structures have been devised to optimize CAR-NK cell functions. For example, a unique CAR-NK cell line with NKG2D-DAP10-CD3ζ structure designed to exploit NKG2D-MICA/B interactions has exhibited remarkable cytotoxicity in acute lymphoblastic leukemia (ALL), osteosarcoma, prostate carcinoma, and rhabdomyosarcoma models (18).
To date, three clinical trials have been conducted to examine PB-CAR-NK cells. Regarding hematological malignancies, one was conducted to explore the efficacy of haploidentical PB-derived anti-CD19-CAR-NK (4-1BB-CD3ζ) in B-lineage ALL patients (NCT00995137), and another was carried out to evaluate the anti-tumor effect of a similar anti-CD19-CAR-NK in patients with refractory ALL (NCT01974479). For solid tumors, a phase I clinical trial is being planned by Guangzhou Medical University in China to explore both the efficacy and safety of autologous/allogeneic broad-spectrum NKG2D-ligand-targeting CAR-NK in multiple metastatic solid tumors (NCT03415100). Considering the exceptional outcomes observed in the corresponding pre-clinical studies, there is anticipation for noteworthy results from these clinical trials (Table 1).
Table 1. Clinical trials and products of CAR-NK cell therapy.
| Sources of NK cells | Trial identifier | Product name | Tumor type | Target | Stage | Status |
| CAR-NK, chimeric antigen receptor-natural killer; PB, peripheral blood; CB, cord blood; NHL, non-Hodgkin lymphoma; ALL, acute lymphoblastic leukemia; AML, acute myeloid leukemia; MDS, myelodysplastic neoplasms; MM, multiple myeloma; BCL, B-cell lymphoma; CLL, chronic lymphocytic leukemia; HER2, human epidermal growth factor receptor 2. *, In this trail, CAR structure refered to novel chimeric costimulatory converting receptor, comprising mainly extracellular domain of PD1, transmembrane and cytoplasmic domains of NKG2D, and cytoplasmic domain of 41BB. | ||||||
| PB-NK | NCT05645601 | JD010 | B-cell malignancies | CD19 | Phase I | Recruiting |
| NCT05410717 | − | Ovarian cancer, testis cancer and other endometrial cancers |
Claudin6 | Phase I/IIa | ||
| NCT05487651 | KUR-502 | B-cell NHL or leukemia | CD19 | Phase I | Recruiting | |
| NCT03692637 | − | Epithelial ovarian cancer | Mesothelin | Early phase I | Unknown status | |
| NCT00995137 | − | B-lineage ALL | CD19 | Phase I | Completed | |
| NCT01974479 | − | B-lineage ALL | CD19 | Phase I | Suspended | |
| NCT03415100 | − | Metastatic solid tumors | NKG2D ligands | Phase I | Unknown status | |
| NCT05020678 | NKX019 | B-cell malignancies | CD19 | Phase I | Recruiting | |
| NCT04623944 | NKX101 | AML or MDS | NKG2D ligands | Phase I | Recruiting | |
| CB-NK | NCT05247957 | − | AML | NKG2D ligands | Phase I | Terminated |
| NCT05472558 | − | B-cell NHL | CD19 | Phase I | Recruiting | |
| NCT04796675 | − | B-cell malignancies | CD19 | Phase I | Recruiting | |
| NCT05922930 | − | Ovarian cancer, mesonephric-like adenocarcinoma, and pancreatic cancer |
TROP2 | Phase I/II | Not yet recruiting | |
| NCT05110742 | − | T-cell malignances | CD5 | Phase I/II | Not yet recruiting | |
| NCT05008536 | − | MM | BCMA | Early phase I | Recruiting | |
| NCT03056339 | − | B-cell malignancies | CD19 | Phase I/II | Completed | |
| NCT05092451 | − | Leukemia, lymphoma, or multiple myeloma | CD70 | Phase I/II | Recruiting | |
| NCT05667155 | − | B-cell NHL | CD19, CD70 | Phase I | Recruiting | |
| NCT05020015 | − | B-cell NHL or indolent NHL | CD19 | Phase II | Recruiting | |
| NCT05703854 | − | Renal cell carcinoma, mesothelioma and osteosarcoma | CD70 | Phase I/II | Recruiting | |
| NCT05842707 | − | B-cell NHL | CD19, CD70 | Phase I/II | Recruiting | |
| NCT05008575 | − | AML | CD33 | Phase I | Recruiting | |
| NK-92 | NCT02944162 | − | AML | CD33 | Phase I/II | Unknown status |
| NCT03940833 | − | MM | BCMA | Phase I/II | Unknown status | |
| NCT02892695 | PCAR-119 | Leukemia, lymphoma | CD19 | Phase I/II | Unknown status | |
| NCT02742727 | − | Leukemia, lymphoma | CD7 | Phase I/II | Unknown status | |
| NCT02839954 | − | Hepatocellular carcinoma, non-small cell lung cancer, pancreatic carcinoma, triple-negative invasive breast carcinoma, malignant glioma of brain, colorectal carcinoma, gastric carcinoma |
MUC1 | Phase I/II | Unknown status | |
| NCT03383978 | − | Glioblastoma | HER2 | Phase I | Recruiting | |
| NCT03656705 | − | Non-small cell lung cancer | −* | Phase I | Completed | |
| iPSC-NK | NCT05182073 | FT576 | MM | BCMA | Phase I | Recruiting |
| NCT04245722 | FT596 | BCL, CLL | CD19 | Phase I | Active, not recruiting | |
| NCT04555811 | FT596 | NHL | CD19 | Phase I | Active, not recruiting | |
| NCT05395052 | FT536 | Non-small cell lung cancer, colorectal cancer, breast cancer, ovarian cancer, pancreatic cancer, head and neck cancer, gastroesophageal cancer | MICA/ MICBα3 |
Phase I | Active, not recruiting | |
| NCT05336409 | CNTY-101 | B-cell malignancies | CD19 | Phase I | Recruiting | |
CB-derived CAR-NK cells
CB-NK cells
CB-NK cells are NK cells that differentiate from hematopoietic stem cells in umbilical CB. CB is an off-the-shelf source of allogeneic NK cells for several reasons. First, the convenient collection and cryopreservation of CB makes it easy to store (19). Second, compared to other sources of grafts, CB has fewer T cells, thereby reducing the risk of GVHD (20). Moreover, CB provides a more stable and higher number of NK cells than PB (21). CB also comprises more NK cell progenitors (22).
Advances in research on CB-CAR-NK
Examination of CB-NK cells has entered clinical trials. A phase I/II clinical trial (NCT03056339) was carried out to test the efficacy and safety of anti-CD19 CAR-CB-NK cells in 11 patients with relapsed or refractory CD19-positive cancers. CB-NK cells were designed to co-express anti-CD19, interleukin (IL)-15, and inducible caspase-9 (iCasp9). IL-15 is a crucial cytokine involved in the proliferation and persistence of NK cells. Suicide genes are safety switches that prevent toxicity. A total of eight (73%) patients presented a response, of which seven had complete remission (CR), and one had remission with respect to Richter’s transformation with persistent CLL. The most striking finding of this study was the complete irrelevance of cytokine release syndrome (CRS)/neurotoxicity/GVHD and CAR-NK therapy (23).
Other phase I clinical studies using CB-CAR-NK cells with a similar CAR structure (anti-CD19) in patients with lymphoma and leukemia have also been launched and are currently underway. Furthermore, the effects and safety of CB-CAR-NK cells in solid cancers are also being studied (NCT05703854 and NCT05922930) (Table 1).
NK cell line-derived CAR-NK cells
NK cell lines
In addition to PB/CB-derived NK cells, there are numerous mature NK cell lines, such as NK-92, NKL, HANK1, IMC-1, SNK-6, and SNT-8, all of which can be cultured on a large scale in vitro to provide sufficient cells for application. Among them, NK-92 is the only NK cell line approved for clinical trials and is an important supplier for the manufacture of CAR-NK (24).
The NK-92 cell line has unique molecular characteristics, such as the expression of CD2+, CD56+, CD3−, and CD16− (25). NK-92 cells are functionally characterized by their high activation and dependence on IL-2 for their cytotoxic killing capabilities. These cells have an exceptional ability to recognize tumor cells and exert potent cytotoxic effects (26). NK-92 cells are equipped with activating receptors and sufficient cytolytic molecules (27,28). NK-92 cells express low levels of inhibitory receptors such as NKG2A to avoid cancer cell-mediated inhibition (25). Antibody-dependent cellular cytotoxicity (ADCC) is another pathway used by NK cells to kill cancer cells. Despite the low expression of CD16 reported in NK-92 cells, NantKwest successfully engineered an NK cell product with a high affinity for CD16 (29). This modification aimed to address this limitation and enhance the cytotoxicity of NK-92 cells. Clinical trials registered under the identifiers NCT03027128 and NCT03853317 are currently being conducted to investigate the effectiveness of this approach. For clinical use, the immortalized NK-92 cell line can be easily cryopreserved in vitro and expanded to large numbers via good manufacturing practices (GMP) (30). According to GMP standards, Tam et al. successfully developed a highly efficient protocol that allowed the proliferation of NK-92 cells more than 200-fold within a 2−2.5-week period. This protocol not only meets clinical immunotherapeutic requirements but also ensures the quality and safety of proliferated cells (31).
Advances in research on CAR-NK-92 therapy
CAR-NK-92 therapy has achieved outstanding results for hematological malignancies and solid tumors. Oelsner et al. constructed a CAR-NK-92 cell line that targeted FMS-like tyrosine kinase 3 (FLT3), which is overexpressed in B-ALL, and confirmed its notable anti-tumor cytotoxicity in SEM B-ALL xenograft models (32). Romanski et al. found that engineered NK-92 cells with an anti-CD19 CAR structure could recover their significant cytotoxicity against CD19-expressing B-precursor leukemia cell lines and lymphocytes from patients with leukemia (33). A phase I/II clinical trial (NCT02944162) reported the efficacy and safety of CAR-NK-92 cells targeting CD33 in three patients with relapsed and refractory acute myeloid leukemia (AML) (34). Clinical trials NCT02892695 exploring the efficacy of 3G CD19-CAR-NK-92 cells and NCT02742727 exploring the safety of CD7-CAR-NK-92 cells in lymphoma and leukemia are still underway.
As for solid tumors, CAR-NK-92 also presented outstanding efficacy. To overcome ovarian cancer, one of the most lethal gynecological cancers, Ao et al. designed anti-folate receptor α (αFR) CAR-NK-92 cells and confirmed their effective killing effect on ovarian cancer cells in vitro and ovarian cancer mouse models (35). Klapdor et al. constructed a novel dual-CAR structure in NK-92 cells (anti-CD24 and anti-mesothelin) that showed high cytotoxicity in both ovarian cancer cell lines and primary ovarian cancer cells (36). Han et al. found that intracranial administration of anti-EGFR CAR-NK-92 cells inhibited tumor growth and prolonged tumor-bearing mouse survival in glioblastoma mouse models (37). Esser et al. showed that CAR-NK-92 cells targeting GD2 showed enhanced cytotoxicity in neuroblastoma pre-clinical models (38). In addition, anti-MUC1 CAR-NK-92 cells were also utilized to treat relapsed or refractory solid tumors (NCT02839954).
In addition to single CAR modifications, promising combination strategies have been used to boost CAR-NK efficacy in solid tumors. Zhang et al. showed the synergistic effects of regorafenib and CAR-NK-92 cells targeting epithelial cell adhesion molecules (EpCAM) in colorectal cancer xenograft mouse models (39). A phase I clinical trial (NCT03383978) targeting glioblastoma with anti-HER2 CAR-NK-92 and another phase I clinical trial (NCT03656705) designed for non-small cell lung cancer were initiated to further explore the efficacy and safety of CAR-NK-92 cells in the treatment of solid clinical tumors (Table 1).
iPSC-derived CAR-NK cells
iPSC-derived NK cells
The development of iPSCs was first initiated by Takahashi using Oct3/4, Sox2, c-Myc, and Klf4 (40). Subsequently, human iPSCs were successfully generated and used as renewable cell sources (41). Similar to embryonic stem cells (ESCs), iPSCs exhibit indefinite proliferation and pluripotency. Successful induction of iPSCs allows for the convenient acquisition of pluripotent stem cells without embryos. Blood cells derived from iPSCs have characteristics similar to those derived from ESCs (42).
A robust system for the production of clinical grade iPSC has been developed (43). For iPSC-CAR-NK products, CAR structural modifications were executed in the undifferentiated state of iPSCs. Subsequently, modified iPSCs are differentiated in a feeder-dependent or feeder-free manner in two steps: First, iPSCs are stimulated by stem cell factor (SCF), vascular endothelial growth factor, and bone morphogenic protein 4 to differentiate into hematopoietic stem cells. Subsequently, CD34+ hematopoietic stem cells are selected to further differentiate into NK cells under stimulation by IL-3, IL-7, IL-15, SCF, and FLT3L (44). Finally, differentiated iPSC-CAR-NK cells were co-cultured with artificial antigen-presenting cells (aAPC and K562 cells expressing mbIL15/mbIL21 + 41BBL) for clinical-scale expansion (45). Zeng et al. established a practical strategy to expand the number of NK cells without KIRs, thereby broadening the range of eligible patients (46). iPSC-derived NK cells display many of the same characteristics as primary NK cells, including the presence of specific markers, such as CD56, NKG2D, and CD16 (47). Recent evidence suggests that iPSC-derived NK cells have comparable or more potent anti-tumor effects than PB/CB NK cells (48). Moreover, iPSC-derived NK cells are more suitable for expressing CAR structures than primary NK cells (44).
Advances in research on iPSC-CAR-NK cells
Pre-clinical and clinical studies of iPSC-derived NK cells have been conducted in recent years (49). A high-affinity, non-cleavable variant of CD16a was introduced and engineered in human iPSCs, resulting in the creation of potent high-affinity noncleavable variant of CD16a (hnCD16)-iPSC-NK cells with improved ADCC properties (50). Furthermore, iPSC-NK cells armed with anti-EpCAM CAR have been developed that show potent cytotoxicity against EpCAM-positive cancer cells that are resistant to NK cell attack (51).
Clinical studies have investigated the effectiveness of iPSC-CAR NK cells against various malignancies. The ELiPSE-1 study (NCT05336409) is currently recruiting participants to evaluate the safety, pharmacokinetics, and preliminary efficacy of CNTY-101 in CD19-positive B cell malignancies. Furthermore, clinical trials using iPSC-CAR-NK cells such as FT576 and FT596 targeting BCMA (NCT05182073) and CD19 (NCT04245722 and NCT04555811), respectively, are underway for examining their therapeutic potential for hematological malignancies. In particular, FT536 is specifically designed to target MICA/MICBα3, which are NKG2D ligands for the treatment of solid cancers (NCT05395052). Due to the novelty of iPSC-CAR NK cells, more clinical experiments are required to explore their safety and efficacy (Table 1).
Challenges and potential solutions for CAR-NK cells derived from different sources
In vitro expansion and cryopreservation of PB-NK cells
Due to the low frequency of NK cells in PB, in vitro expansion is necessary to obtain clinical-scale quantities (52). Feeder-dependent and feeder-free approaches are the two main expansion approaches.
Co-stimulatory signaling is essential for NK cell expansion. Feeder cells can provide different co-stimulatory signals to NK cells via cell-to-cell contact, allowing clinical-scale expansion of NK cells (53). K562, an erythroleukemia cell line lacking HLA antigen expression, was initially found to be co-stimulatory. Irradiated K562 cells have been widely used to enhance NK cell expansion (54). K562, which was modified to express specific co-stimulatory molecules, exhibited better NK-expansion efficacy in clinical trials. For example, K562-mb15-4-1BBL is capable of increasing NK expansion and upregulating NK activation markers such as CD69, CD25, NKG2D, NKp30, NKp44, and NKp46. No T cell expansion, genetic alterations, or potential oncogenic effects has been observed (17,55). MbIL-21 is an excellent substitute for mbIL-15. K562 cells expressing mbIL-21 showed enhanced expansion of NK cells (>47,000-folds in 21 d), and these proliferated cells always exhibited increased activation and purity without senescence, represented by telomere shortening (56). The irradiated EBV-transformed lymphoblastoid cell line has also been used to obtain clinical-scale NK cells, and these expanded NK cells exhibit superior activation and cytotoxicity compared to naïve NK cells (57,58).
To further strengthen the safety of feeder-dependent expansion systems, many non-tumor or non-genetically modified feeder cells have been explored. Two phase I clinical trials examining adoptive NK therapy used RetroNectin-stimulated T cells (RN-T) to expand autologous NK cells, and both obtained sufficient NK cells with high purity, functionality (high expression of NKG2D and CD16), and safety (59,60). Irradiated autologous PBMCs are alternate choices (61), and their combination with anti-CD16 monoclonal antibodies was confirmed to be an effective approach to expand highly purified cytotoxic NK cells with potent anti-tumor function both in vitro and in vivo (61,62). Compared to feeder cells, feeder-free approaches may facilitate easier licensing as a clinical-grade expansion platform, regardless of the infusion of viable feeder cells. Based on the existing application of different non-cell-based activating supplements (e.g., cytokines) for clinical-grade NK cell expansion, optimization of the combination, adjusting sequencing, and duration of exposure time will be more extensively examined in the future (63,64).
PB-NK cells exhibit reduced cytotoxicity after cryopreservation and long-distance transportation; however, this can be partially overcome by the expansion process, which largely limits the therapeutic effect of adoptive NK therapy (65). Therefore, the exploration of effective cryoprotective agents (CPAs) is a promising strategy. Dimethyl sulfoxide (DMSO), a widely used cryopreservation reagent, exhibits broad toxicity. It is capable of altering the expression of NK cell markers and impairing NK in vivo functions (66). In view of this, Yao et al. designed a biocompatible chitosan-based nanoparticle, which can mediate efficient DMSO-free NK-cell cryopreservation via intracellular delivery of non-DMSO CPAs. More importantly, NK cells cryopreserved in this manner maintain their strong cytotoxicity, degranulation, and cytokine production functions (67).
Simultaneous maturation and expansion of CB-NK cells
The use of CB-NK cells presents two significant challenges for investigators: the limited number of NK cells in a CB unit and their naïve phenotype. Although the percentage of NK cells was similar or even higher in CB than in PB, the total number of NK cells was limited by the small volume of a single CB unit (68). In addition, CB-NK cells are less mature than their PB counterparts (69). CB-NK cells exhibit a different phenotype characterized by decreased expression of molecules such as CD16 and granzyme B and increased expression of NKG2A and CXCR3 (68,70). The immature phenotype observed in umbilical CB-NK cells is indicative of reduced cytotoxicity and increased homing propensity. Consequently, CB-NK cells generally demonstrate lower anti-cancer activity than PB-NK cells. Therefore, any attempt to expand CB-NK cells must be accompanied by an effort to enhance their activation.
The simultaneous resolution of insufficient in vitro expansion and immature killing capacity may be achieved via the use of expansion techniques, which commonly exploit the benefits of interleukins, particularly IL-2 and IL-15 (71). Through ex vivo expansion technology, the number of NK cells can be substantially augmented, with an approximately (1,800−2,400)-fold increase observed in fresh or cryopreserved CB units (53). Furthermore, the activation and maturation of NK cells, contributing to their effective killing abilities, are achieved via the influence of cytokines (72). CB-NK cells can also be activated by feeder cells. Liu et al. successfully created universal antigen-presenting cells (uAPC) by engineering K562 cells with increased expression of CD48, 4-1BBL, and mbIL21 to promote the expansion and activation of CB-CAR-NK cells (73).
In vivo persistence of NK92 cells
Despite the advantages of NK-92 cells over PB/CB-NK cells in terms of expansion and modification, they still face challenges due to their limited persistence in vivo. There are two possible reasons for the limited persistence of NK-92 cells. First, for lymphoma, NK-92 cells must be irradiated before adoptive transfer to eliminate the risk of tumorigenicity and Epstein-Barr viral susceptibility (74). However, irradiation was detrimental to the persistence of NK-92 cells in vivo. Furthermore, the viability and proliferation of NK-92 cells are highly dependent on a continuous supply of low-dose IL-2. In the absence of IL-2, NK-92 cells exhibited a rapid decline in survival, leading to death in 72 h. Therefore, when introduced into the human body, the absence of IL-2 poses a significant threat to the longevity of NK-92 cells (25).
Enhancing the persistence of NK-92 cells can be achieved via gene modifications, particularly by targeting various elements, such as cytokines. For example, NK-92 cells can be modified by attaching IL-2 to their plasma membranes to stimulate nearby IL-2 receptors. This membrane-bound protein (MBP) NK-92 cells exhibited exogenous IL-2 independent proliferation and showed improved infiltration and persistent activity in an A549 spheroid solid tumor model (75). Similarly, mbIL-2-expressing NK-92 cells showed improved persistence and enhanced anti-tumor activity in a leukemia xenograft model (76). IL-15 is another cytokine commonly used in laboratory and clinical investigations to sustain survival and enhance the cytotoxicity of NK cells. NK-92 cells co-expressing IL-15 and EpCAM/breast cancer-specific CAR showed persistent and increased cytotoxicity against EpCAM-expressing breast carcinoma cells in vitro (77).
In addition to cytokines, other elements can be modified to optimize the persistence of NK-92 cells. For example, in vivo studies have shown that NK-92 cells expressing erythropoietin or thrombopoietin receptors have a longer lifespan (78). Similarly, NK-92 cells with truncated PD-1 (tPD-1) exhibit enhanced persistence compared to standard NK-92 cells, as tPD-1 blocks the PD-1/PD-L1 inhibition (79).
Common challenges associated with CAR-NK cells
Gene modification of CAR-NK products
Overcoming the resistance of NK cells to transduction is a significant hurdle in the successful and efficient gene modification of CAR-NK cells. Compared to T cells, NK cells show a higher sensitivity to foreign genetic materials and a lower transduction efficiency (80). Mechanically, pattern recognition receptors in NK cells recognize foreign genetic materials and trigger an innate defense against them (81). Therefore, the potential risks of insertional gene mutations should not be overlooked. In the presence of high titers of vital vectors, NK cells encounter the potential genotoxicity associated with insertional mutations (82).
Currently, transfection methods are divided into two types: viral and non-viral. Viral vectors have emerged as highly efficient and superior gene modification tools compared to non-viral alternatives (83). As an important component of viral transfection systems, lentiviral transfection has been used in phase I/II clinical research on CAR NK cell therapy for hematological (NCT05472558) and solid (NCT05410717) malignancies. Other retroviral vectors are also primary choices to modify NK cell genes. For example, Suerth et al. confirmed that alpha-retroviral vectors are useful tools for the application of NK cell-mediated cancer immunotherapy (84).
To date, many efforts have been made to enhance the transduction efficiency. These include the implementation of multiple rounds of transduction, co-culture techniques involving feeder cells, and the development and refinement of novel vector designs.
(1) Multiple rounds of transduction
Adopting multiple rounds of transduction represents a direct approach to improving the efficiency of gene expression. Guven et al. observed that the percentage of NK cells transduced on d 21 was higher in cells that underwent two rounds of transduction than in cells that underwent a single round of transduction (75.4% vs. 51.9%) (85). In addition, in the presence of the lentiviral transduction enhancer dextran, the transduction rate of NK cells reached 40% after one round of transduction and increased to 100% after two rounds (86). Therefore, repeated transduction may be a potential method to increase transduction efficiency.
(2) Co-culture with feeder cells
Feeder cells can be used to improve the production of CAR-NK cells. Despite their potential impact on viral transfection efficiency, feeder cells effectively enhance the recovery and expansion of transfected NK cells. Allan et al. investigated the delicate balance between gene editing efficiency and expansion potential and showed that the ideal pre-activation duration of feeder cells is 5−7 d prior to the lentiviral transduction (87). EBV-transformed lymphoblastoid cell lines and genetically modified K562 cells are feeder cells that are commonly used to achieve clinical-scale NK cell proliferation (23,88). Mechanically, the secretion of IL-2 by feeder cells serves as a highly effective enhancer of NK cell expansion and transduction (57). Furthermore, physical contact between feeder cells and NK cells, as well as the binding of ligands on the feeder cells (e.g., 4-1BBL (CD137L), mbIL-15, and mbIL-21) to their corresponding stimulatory receptors on NK cells, is crucial for successful expansion of NK cells (89,90).
(3) Vector and transduction enhancer
The design of new vectors appears to be another feasible method. One such example is the use of baboon envelope-pseudotyped lentiviral vectors, which have been developed to promote viral entry. Colamartino et al. demonstrated that these vectors achieved a transduction efficiency of 38.3%±23.8% in NK cells, which increased to 58.4%±7.8% after re-expansion (91).
The use of transduction enhancers, such as dextran and IL-2, is another option. Müller et al. conducted a comparative study of two different retroviral vector platforms with the addition of retronectin and vectofusin-1. They found that the combination of RD114-TR pseudotyped retroviral and vectofusin-1 was the optimal strategy to engineer PB-derived NK cells (92).
(4) Non-viral gene modification
Considering the restricted vector capacity and potential risk of tumorigenicity associated with viral transfection, various non-viral vectors have been extensively investigated in recent years to overcome these limitations. Electroporation, nucleofection, and lipofection are the most commonly used non-viral transduction methods (93). Compared to lipofection, electroporation has shown greater efficiency and suitability for clinical translation, although it may be less cost-effective (94). mRNAs are the main cargo involved in non-viral transduction. Carlsten et al. used electroporation to deliver the mRNA of high-affinity CD16 and the chemokine receptor C-C motif chemokine receptor 7 to primary NK cells. This resulted in 95% of NK cells successfully expressing the target mRNA. The engineered cells exhibit enhanced migration and cytotoxicity against antibody-coated lymphoma cells (95). However, while mRNA has a lower risk of insertional mutation as it cannot integrate into DNA, it poses a limitation for non-viral transduction methods in achieving permanent CAR structure expression. Additionally, the limited capacity of mRNA as a cargo hinders the development of non-viral transduction (96).
Various strategies have been explored to overcome the aforementioned hurdles, such as the utilization of DNA transposons, CRISPR/Cas9 technology, and nanoparticles. DNA transposons have become popular non-viral gene delivery tools for achieving stable gene expression (97). Transposon-mediated CAR transduction, especially with piggyBac and sleeping beauty transposon systems, has shown promising efficacy in achieving stable CAR expression in NK cells through a close-to-random profile of genomic integration (98). Another promising approach is the combination of non-viral gene delivery methods with CRISPR delivery systems (99). Specifically, Nguyen et al. electroporated Cas9 ribonucleoproteins in NK cells, which improved the transfection efficiency. Moreover, multifunctional nanoparticles (MF-NPs) can be used to genetically express anti-EGFR-CAR structures on NK cells and image MF-NP-loaded NK cells in vivo (100).
Challenges of CAR-NK cells in solid tumor treatment
CAR-NK cells from different sources have limited effectiveness in solid tumor treatment and yield unsatisfactory outcomes. This can be attributed to several factors. First, decreased expression of chemokine ligands, such as CXCL1 and CXCL9, impairs NK cell trafficking ability, restricting their infiltration into tumor sites (101). For example, in hepatocellular carcinoma, miR-561-5p overexpression downregulates CXCL1 expression and blocks CXCR1+NK cell infiltration (102). Furthermore, the immunosuppressive characteristics and the abnormal metabolic profile of tumor microenvironment (TME) hinder the survival and function of NK cells (103,104). Fortunately, various strategies have shown promise in addressing these challenges in pre-clinical tumor models.
(1) Enhancing infiltration of CAR-NK cells
One approach to enhance the infiltration of NK cells into tumor sites is to increase the expression of chemokines in NK cells and stimulate cancer cells to secrete the corresponding chemokine ligands. This can be achieved by various methods. First, NK cells can be designed to co-express a CAR structure and chemokine receptors, such as CXCR3 and CXCR4, which are key mediators of NK cell trafficking. Müller et al. successfully enhanced tumor infiltration by NK cells by designing novel NK cells with EGFRvIII-specific CAR and CXCR4 (105). Similarly, CXCR1-engineered NK cells exhibited increased infiltration of human tumors in a xenograft model (106). Second, certain anti-tumor agents, such as radiotherapy and autophagy inhibitors, can disrupt normal tumor processes, leading to increased secretion of chemokine ligands (107,108). For example, dipeptidyl peptidase inhibitors and curaxins have been shown to enhance the secretion of CXCL9 and CXCL10 in different solid tumors, promoting the infiltration of CXCR3+ NK cells into the TME (109). In addition, nanoparticles can facilitate NK cell infiltration. For instance, magnetic nanoparticles loaded with NK-92 cells showed a 17-fold increase in infiltration when a magnetic field was applied compared to normal NK-92 cells (110).
(2) Optimization of CAR-NK cells
To improve the function of NK cells, it is important to directly modify NK cells to overcome unfavorable conditions in the TME. One promising approach is to adapt and modulate CAR-NK cells to respond effectively to the components of TME. Therefore, reengineering the CAR structure has shown great potential. A unique CAR, namely the novel chimeric costimulatory converting receptor (CCCR), was designed to address this challenge. CCCR incorporates the domain of PD1 (extracellular), NKG2D (transmembrane and cytoplasmic), and 41BB (cytoplasmic). This design aimed to convert the inhibitory PD-1 signal into an activating one (111). When expressed in NK-92 cells, CCCR-NK-92 cells were able to overcome immunosuppressive TME and exhibited enhanced anti-tumor efficacy.
Memory-like NK cells have increased production of IFN-γ, which is related to the upregulation of killing-activating receptors and downregulation of killing inhibitory receptors. This ultimately leads to a superior killing effect compared to common NK cells (112). Inducing immunological memory of NK cells is an alternative method to enhance NK cell activity in solid tumors (113). For example, Chang et al. developed PD-L1-induced memory-like iPSC-NK cells that respond to PD-L1 signaling, resulting in enhanced proliferation and cytotoxicity. In a mouse xenograft model, these cells exhibited higher anti-tumor cytotoxicity than other CAR-NK cells (114).
Additionally, the effectiveness of NK cells with different CAR structures may vary depending on the type of cancer. For example, in prostate cancer mouse models, anti-PSCA-DAP12 CAR-NK cells showed greater cytotoxicity compared to anti-PSCA-CD3ζ CAR-NK cells (115). Similarly, Li et al. evaluated different CAR structures in iPSC-NK cells and found that NKG2D-2B4-CD3ζ CAR exhibited the strongest activating effect on NK cells in an ovarian cancer model (98). Therefore, it is crucial to compare and optimize various CARs for different types of cancer to maximize the cytotoxic effects of CAR-NK cells.
(3) TME modification
In addition, it is important to regulate the immune and metabolic conditions in the TME to enhance the effectiveness of CAR-NK cells. Various agents, such as chemotherapy and immune checkpoint inhibitors (ICIs), can modulate the immune response in the TME and improve the persistence and cytotoxicity of NK cells (116,117). Chemotherapy can directly activate NK cells and modify immune response in the TME to support NK cell functions. It achieves this by improving the immunogenicity of cancer cells and regulating the recruitment and function of immunosuppressive cells (118). For example, Klapdor et al. found that treating CD44-positive ovarian cancer cells with anti-CD44 NK cells and cisplatin simultaneously improved the anti-tumor capacity of NK cells (119).
ICIs are effective in regulating immune interactions between NK and tumor cells. In solid cancers, prominent inhibitory checkpoints, such as PD-1, NKG2A, and KIRs, can inhibit NK cell activation (120,121). Various antibodies have been developed to block these inhibitory checkpoints and restore CAR-NK cell function to overcome immune inhibition (122). Wang et al. showed that combining anti-PSMA CAR NK-92 cells with an anti-PD-L1 antibody improves anti-tumor efficacy in a castration-resistant prostate cancer mouse model (123). Furthermore, clinical trials have confirmed that monalizumab, an anti-NKG2A monoclonal antibody, improves NK cell activity in solid cancers (124). The combination of monalizumab with CAR-NK cell therapy has the potential to enhance its therapeutic effects.
Additionally, abnormal metabolic conditions in TME, such as hypoxia, nutrient deprivation, and accumulation of metabolites (e.g., reactive oxygen species and adenosine), have a significant effect on the metabolism and energy supply of NK cells, ultimately affecting their anti-tumor properties (125,126). Therefore, modulating metabolism can be a promising method to enhance the performance of NK cells in TME. For example, combining NKG2D-specific CAR-NK-92 cells with CD73 blockade improves therapeutic efficacy in CD73+ human lung cancer models (127). CD73 is associated with the accumulation of adenosine, a metabolite with immunosuppressive effects that leads to the exhaustion of NK cells (128) (Figure 2).
Figure 2.
Individual and common challenges faced by CAR-NK cells derived from different sources. Common challenges: Left, NK cells are initially resistant to transfection during gene modification; Right, the efficacy of CAR-NK therapy in solid tumors is still limited. Individual challenges: The efficiency of expansion needs to be improved to meet clinical requirements in both PB- and CB-derived CAR-NK cells. More advanced techniques are required to maintain the activity of PB-CAR-NK cells during cryopreservation. Compared to PB-NK cells, CB-NK cells are less mature. Preliminary irradiation and reliance on IL-2 leads to limited in vivo persistence of CAR-NK-92 cells. The clinical safety and efficacy of iPSC-CAR-NK cells require further verification. CAR-NK, chimeric antigen receptor-natural killer; PB, peripheral blood; CB, cord blood. The figure is created with BioRender.com.
Conclusions and perspective
CAR-NK cells derived from PB, CB, and NK-92 cell lines and iPSCs are effective in anti-tumor therapy. The abundant allogeneic suppliers of NK cells undoubtedly provide a promising opportunity for clinical-scale manufacturing and application of CAR-NK therapy. However, each source also has its problems that need to be solved and is associated with common challenges that impede effective CAR-NK production. Although CAR-NK therapy has shown outstanding anti-tumor effects on pre-clinical studies of both hematological and solid tumors, corresponding clinical trials with CAR-NK cells are just beginning, and many critical results have not yet been published. Therefore, to obtain reliable clinical-level off-the-shelf CAR-NK products, follow-up comparisons and verification of the clinical efficacy and safety of CAR-NK cells from different sources and optimization and standardization of CAR-NK manufacturing processes are required.
Acknowledgements
None.
References
- 1.June CH, O’Connor RS, Kawalekar OU, et al CAR T cell immunotherapy for human cancer. Science. 2018;359:1361–5. doi: 10.1126/science.aar6711. [DOI] [PubMed] [Google Scholar]
- 2.Xie G, Dong H, Liang Y, et al CAR-NK cells: A promising cellular immunotherapy for cancer. EBioMedicine. 2020;59:102975. doi: 10.1016/j.ebiom.2020.102975. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 3.Peng X, Chen L, Chen L, et al Chimeric antigen receptor-natural killer cells: Novel insight into immunotherapy for solid tumors (Review) Exp Ther Med. 2021;21:340. doi: 10.3892/etm.2021.9771. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 4.Karadimitris A Cord blood CAR-NK cells: Favorable initial efficacy and toxicity but durability of clinical responses not yet clear. Cancer cell. 2020;37:426–7. doi: 10.1016/j.ccell.2020.03.018. [DOI] [PubMed] [Google Scholar]
- 5.Siegler EL, Zhu Y, Wang P, et al Off-the-shelf CAR-NK cells for cancer immunotherapy. Cell Stem Cell. 2018;23:160–1. doi: 10.1016/j.stem.2018.07.007. [DOI] [PubMed] [Google Scholar]
- 6.Ettinghausen SE, Moore JG, White DE, et al Hematologic effects of immunotherapy with lymphokine-activated killer cells and recombinant interleukin-2 in cancer patients. Blood. 1987;69:1654–60. doi: 10.1182/blood.V69.6.1654.1654. [DOI] [PubMed] [Google Scholar]
- 7.Liu X, Ran R, Shao B, et al Combined peripheral natural killer cell and circulating tumor cell enumeration enhance prognostic efficiency in patients with metastatic triple-negative breast cancer. Chin J Cancer Res. 2018;30:315–26. doi: 10.21147/j.issn.1000-9604.2018.03.04. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 8.Cooper MA, Fehniger TA, Caligiuri MA The biology of human natural killer-cell subsets. Trends Immunol. 2001;22:633–40. doi: 10.1016/s1471-4906(01)02060-9. [DOI] [PubMed] [Google Scholar]
- 9.Romagnani C, Juelke K, Falco M, et al CD56brightCD16- killer Ig-like receptor- NK cells display longer telomeres and acquire features of CD56dim NK cells upon activation. J Immunol. 2007;178:4947–55. doi: 10.4049/jimmunol.178.8.4947. [DOI] [PubMed] [Google Scholar]
- 10.Domaica CI, Fuertes MB, Uriarte I, et al Human natural killer cell maturation defect supports in vivo CD56(bright) to CD56(dim) lineage development. PloS One. 2012;7:e51677. doi: 10.1371/journal.pone.0051677. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 11.Parkhurst MR, Riley JP, Dudley ME, et al Adoptive transfer of autologous natural killer cells leads to high levels of circulating natural killer cells but does not mediate tumor regression. Clin Cancer Res. 2011;17:6287–97. doi: 10.1158/1078-0432.Ccr-11-1347. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 12.Carrega P, Pezzino G, Queirolo P, et al Susceptibility of human melanoma cells to autologous natural killer (NK) cell killing: HLA-related effector mechanisms and role of unlicensed NK cells. PLoS One. 2009;4:e8132. doi: 10.1371/journal.pone.0008132. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 13.Morvan M, David G, Sébille V, et al Autologous and allogeneic HLA KIR ligand environments and activating KIR control KIR NK-cell functions. Eur J Immunol. 2008;38:3474–86. doi: 10.1002/eji.200838407. [DOI] [PubMed] [Google Scholar]
- 14.Koepsell SA, Miller JS, McKenna DH Jr Natural killer cells: a review of manufacturing and clinical utility. Transfusion. 2013;53:404–10. doi: 10.1111/j.1537-2995.2012.03724.x. [DOI] [PubMed] [Google Scholar]
- 15.Borah A, Zhang J, Guo F, et al Immune evasion and therapeutic opportunities based on natural killer cells. Chin J Cancer Res. 2023;35:283–98. doi: 10.21147/j.issn.1000-9604.2023.03.07. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 16.Kruschinski A, Moosmann A, Poschke I, et al Engineering antigen-specific primary human NK cells against HER-2 positive carcinomas. Proc Natl Acad Sci USA. 2008;105:17481–6. doi: 10.1073/pnas.0804788105. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 17.Imai C, Iwamoto S, Campana D Genetic modification of primary natural killer cells overcomes inhibitory signals and induces specific killing of leukemic cells. Blood. 2005;106:376–83. doi: 10.1182/blood-2004-12-4797. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 18.Chang YH, Connolly J, Shimasaki N, et al A chimeric receptor with NKG2D specificity enhances natural killer cell activation and killing of tumor cells. Cancer Res. 2013;73:1777–86. doi: 10.1158/0008-5472.CAN-12-3558. [DOI] [PubMed] [Google Scholar]
- 19.Munoz J, Shah N, Rezvani K, et al Concise review: umbilical cord blood transplantation: past, present, and future. Stem Cells Transl Med. 2014;3:1435–43. doi: 10.5966/sctm.2014-0151. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 20.Knorr DA, Bachanova V, Verneris MR, et al Clinical utility of natural killer cells in cancer therapy and transplantation. Semin Immunol. 2014;26:161–72. doi: 10.1016/j.smim.2014.02.002. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 21.Lin SJ, Kuo ML Cytotoxic function of umbilical cord blood natural killer cells: relevance to adoptive immunotherapy. Pediatr Hematol Oncol. 2011;28:640–6. doi: 10.3109/08880018.2011.613092. [DOI] [PubMed] [Google Scholar]
- 22.Rutella S, Bonanno G, Marone M, et al Identification of a novel subpopulation of human cord blood CD34-CD133-CD7-CD45+lineage- cells capable of lymphoid/NK cell differentiation after in vitro exposure to IL-15. J Immunol. 2003;171:2977–88. doi: 10.4049/jimmunol.171.6.2977. [DOI] [PubMed] [Google Scholar]
- 23.Liu E, Marin D, Banerjee P, et al Use of CAR-transduced natural killer cells in CD19-positive lymphoid tumors. N Engl J Med. 2020;382:545–53. doi: 10.1056/NEJMoa1910607. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 24.Zhang J, Zheng H, Diao Y Natural killer cells and current applications of chimeric antigen receptor-modified NK-92 cells in tumor immunotherapy. Int J Mol Sci. 2019;20:317. doi: 10.3390/ijms20020317. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 25.Gong J, Maki G, Klingemann HG Characterization of a human cell line (NK-92) with phenotypical and functional characteristics of activated natural killer cells. Leukemia. 1994;8:652–8. [PubMed] [Google Scholar]
- 26.Klingemann H, Boissel L, Toneguzzo F Natural killer cells for immunotherapy — Advantages of the NK-92 cell line over blood NK cells. Front Immunol. 2016;7:91. doi: 10.3389/fimmu.2016.00091. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 27.Boissel L, Betancur-Boissel M, Lu W, et al Retargeting NK-92 cells by means of CD19- and CD20-specific chimeric antigen receptors compares favorably with antibody-dependent cellular cytotoxicity. Oncoimmunology. 2013;2:e26527. doi: 10.4161/onci.26527. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 28.Maki G, Klingemann HG, Martinson JA, et al Factors regulating the cytotoxic activity of the human natural killer cell line, NK-92. J Hematother Stem Cell Res. 2001;10:369–83. doi: 10.1089/152581601750288975. [DOI] [PubMed] [Google Scholar]
- 29.Klingemann H The NK-92 cell line-30 years later: its impact on natural killer cell research and treatment of cancer. Cytotherapy. 2023;25:451–7. doi: 10.1016/j.jcyt.2022.12.003. [DOI] [PubMed] [Google Scholar]
- 30.Pasley S, Zylberberg C, Matosevic S Natural killer-92 cells maintain cytotoxic activity after long-term cryopreservation in novel DMSO-free media. Immunol Lett. 2017;192:35–41. doi: 10.1016/j.imlet.2017.09.012. [DOI] [PubMed] [Google Scholar]
- 31.Tam YK, Martinson JA, Doligosa K, et al Ex vivo expansion of the highly cytotoxic human natural killer-92 cell-line under current good manufacturing practice conditions for clinical adoptive cellular immunotherapy. Cytotherapy. 2003;5:259–72. doi: 10.1080/14653240310001523. [DOI] [PubMed] [Google Scholar]
- 32.Oelsner S, Waldmann A, Billmeier A, et al Genetically engineered CAR NK cells display selective cytotoxicity against FLT3-positive B-ALL and inhibit in vivo leukemia growth. Int J Cancer. 2019;145:1935–45. doi: 10.1002/ijc.32269. [DOI] [PubMed] [Google Scholar]
- 33.Romanski A, Uherek C, Bug G, et al CD19-CAR engineered NK-92 cells are sufficient to overcome NK cell resistance in B-cell malignancies. J Cell Mol Med. 2016;20:1287–94. doi: 10.1111/jcmm.12810. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 34.Tang X, Yang L, Li Z, et al First-in-man clinical trial of CAR NK-92 cells: safety test of CD33-CAR NK-92 cells in patients with relapsed and refractory acute myeloid leukemia. Am J Cancer Res. 2018;8:1083–9. [PMC free article] [PubMed] [Google Scholar]
- 35.Ao X, Yang Y, Li W, et al Anti-αFR CAR-engineered NK-92 cells display potent cytotoxicity against αFR-positive ovarian cancer. J Immunother. 2019;42:284–96. doi: 10.1097/cji.0000000000000286. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 36.Klapdor R, Wang S, Morgan M, et al Characterization of a novel third-generation anti-CD24-CAR against ovarian cancer. Int J Mol Sci. 2019;20:660. doi: 10.3390/ijms20030660. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 37.Han J, Chu J, Keung Chan W, et al CAR-engineered NK cells targeting wild-type EGFR and EGFRvIII enhance killing of glioblastoma and patient-derived glioblastoma stem cells. Sci Rep. 2015;5:11483. doi: 10.1038/srep11483. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 38.Esser R, Müller T, Stefes D, et al NK cells engineered to express a GD2-specific antigen receptor display built-in ADCC-like activity against tumour cells of neuroectodermal origin. J Cell Mol Med. 2012;16:569–81. doi: 10.1111/j.1582-4934.2011.01343.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 39.Zhang Q, Zhang H, Ding J, et al Combination therapy with EpCAM-CAR-NK-92 cells and regorafenib against human colorectal cancer models. J Immunol Res. 2018;2018:4263520. doi: 10.1155/2018/4263520. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 40.Takahashi K, Yamanaka S Induction of pluripotent stem cells from mouse embryonic and adult fibroblast cultures by defined factors. Cell. 2006;126:663–76. doi: 10.1016/j.cell.2006.07.024. [DOI] [PubMed] [Google Scholar]
- 41.Takahashi K, Tanabe K, Ohnuki M, et al Induction of pluripotent stem cells from adult human fibroblasts by defined factors. Cell. 2007;131:861–72. doi: 10.1016/j.cell.2007.11.019. [DOI] [PubMed] [Google Scholar]
- 42.Choi KD, Yu J, Smuga-Otto K, et al Hematopoietic and endothelial differentiation of human induced pluripotent stem cells. Stem Cells. 2009;27:559–67. doi: 10.1634/stemcells.2008-0922. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 43.Valamehr B, Robinson M, Abujarour R, et al Platform for induction and maintenance of transgene-free hiPSCs resembling ground state pluripotent stem cells. Stem Cell Reports. 2014;2:366–81. doi: 10.1016/j.stemcr.2014.01.014. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 44.Knorr DA, Ni Z, Hermanson D, et al Clinical-scale derivation of natural killer cells from human pluripotent stem cells for cancer therapy. Stem Cells Transl Med. 2013;2:274–83. doi: 10.5966/sctm.2012-0084. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 45.Zhu H, Kaufman DS An improved method to produce clinical-scale natural killer cells from human pluripotent stem cells. Methods Mol Biol. 2019;2048:107–19. doi: 10.1007/978-1-4939-9728-2_12. [DOI] [PubMed] [Google Scholar]
- 46.Zeng J, Tang SY, Toh LL, et al Generation of “Off-the-Shelf” natural killer cells from peripheral blood cell-derived induced pluripotent stem cells. Stem Cell Reports. 2017;9:1796–812. doi: 10.1016/j.stemcr.2017.10.020. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 47.Goldenson BH, Hor P, Kaufman DS iPSC-derived natural killer cell therapies — expansion and targeting. Front Immunol. 2022;13:841107. doi: 10.3389/fimmu.2022.841107. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 48.Hermanson DL, Bendzick L, Pribyl L, et al Induced pluripotent stem cell-derived natural killer cells for treatment of ovarian cancer. Stem Cells. 2016;34:93–101. doi: 10.1002/stem.2230. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 49.Saetersmoen ML, Hammer Q, Valamehr B, et al Off-the-shelf cell therapy with induced pluripotent stem cell-derived natural killer cells. Semin Immnopathol. 2019;41:59–68. doi: 10.1007/s00281-018-0721-x. [DOI] [PubMed] [Google Scholar]
- 50.Zhu H, Blum RH, Bjordahl R, et al Pluripotent stem cell-derived NK cells with high-affinity noncleavable CD16a mediate improved antitumor activity. Blood. 2020;135:399–410. doi: 10.1182/blood.2019000621. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 51.Tang SY, Zha S, Du Z, et al Targeted integration of EpCAM-specific CAR in human induced pluripotent stem cells and their differentiation into NK cells. Stem Cell Res Ther. 2021;12:580. doi: 10.1186/s13287-021-02648-4. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 52.Fang F, Wang W, Chen M, et al Technical advances in NK cell-based cellular immunotherapy. Cancer Bio Med. 2019;16:647–54. doi: 10.20892/j.issn.2095-3941.2019.0187. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 53.Kundu S, Gurney M, O’Dwyer M Generating natural killer cells for adoptive transfer: expanding horizons. Cytotherapy. 2021;23:559–66. doi: 10.1016/j.jcyt.2020.12.002. [DOI] [PubMed] [Google Scholar]
- 54.Robertson MJ, Cameron C, Lazo S, et al Costimulation of human natural killer cell proliferation: role of accessory cytokines and cell contact-dependent signals. Nat Immun. 1996;15:213–26. [PubMed] [Google Scholar]
- 55.Fernández L, Leivas A, Valentín J, et al How do we manufacture clinical-grade interleukin-15-stimulated natural killer cell products for cancer treatment. Transfusion. 2018;58:1340–7. doi: 10.1111/trf.14573. [DOI] [PubMed] [Google Scholar]
- 56.Somanchi SS, Lee DA Ex vivo expansion of human NK cells using K562 engineered to express membrane bound IL21. Methods Mol Biol. 2016;1441:175–93. doi: 10.1007/978-1-4939-3684-7_15. [DOI] [PubMed] [Google Scholar]
- 57.Granzin M, Stojanovic A, Miller M, et al Highly efficient IL-21 and feeder cell-driven ex vivo expansion of human NK cells with therapeutic activity in a xenograft mouse model of melanoma. Oncoimmunology. 2016;5:e1219007. doi: 10.1080/2162402x.2016.1219007. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 58.Granzin M, Soltenborn S, Müller S, et al Fully automated expansion and activation of clinical-grade natural killer cells for adoptive immunotherapy. Cytotherapy. 2015;17:621–32. doi: 10.1016/j.jcyt.2015.03.611. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 59.Sakamoto N, Ishikawa T, Kokura S, et al Phase I clinical trial of autologous NK cell therapy using novel expansion method in patients with advanced digestive cancer. J Trans Med. 2015;13:277. doi: 10.1186/s12967-015-0632-8. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 60.Ishikawa T, Okayama T, Sakamoto N, et al Phase I clinical trial of adoptive transfer of expanded natural killer cells in combination with IgG1 antibody in patients with gastric or colorectal cancer. Int J Cancer. 2018;142:2599–609. doi: 10.1002/ijc.31285. [DOI] [PubMed] [Google Scholar]
- 61.Parkhurst MR, Riley JP, Dudley ME, et al Adoptive transfer of autologous natural killer cells leads to high levels of circulating natural killer cells but does not mediate tumor regression. Clin Cancer Res. 2011;17:6287–97. doi: 10.1158/1078-0432.CCR-11-1347. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 62.Lee HR, Son CH, Koh EK, et al Expansion of cytotoxic natural killer cells using irradiated autologous peripheral blood mononuclear cells and anti-CD16 antibody. Sci Rep. 2017;7:11075. doi: 10.1038/s41598-017-09259-1. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 63.Alici E, Sutlu T, Björkstrand B, et al Autologous antitumor activity by NK cells expanded from myeloma patients using GMP-compliant components. Blood. 2008;111:3155–62. doi: 10.1182/blood-2007-09-110312. [DOI] [PubMed] [Google Scholar]
- 64.Johnson C, Jesuraj NJ Seeding density is critical for optimizing scalable feeder-free natural killer cell expansion. Cytotherapy. 2020;22:S126. doi: 10.1016/j.jcyt.2020.03.240. [DOI] [Google Scholar]
- 65.Damodharan SN, Walker KL, Forsberg MH, et al Analysis of ex vivo expanded and activated clinical-grade human NK cells after cryopreservation. Cytotherapy. 2020;22:450–7. doi: 10.1016/j.jcyt.2020.05.001. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 66.Yao X, Matosevic S Cryopreservation of NK and T cells without DMSO for adoptive cell-based immunotherapy. BioDrugs. 2021;35:529–45. doi: 10.1007/s40259-021-00494-7. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 67.Yao X, Jovevski JJ, Todd MF, et al Nanoparticle-mediated intracellular protection of natural killer cells avoids cryoinjury and retains potent antitumor functions. Adv Sci (Weinh) 2020;7:1902938. doi: 10.1002/advs.201902938. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 68.Luevano M, Daryouzeh M, Alnabhan R, et al The unique profile of cord blood natural killer cells balances incomplete maturation and effective killing function upon activation. Hum Immunol. 2012;73:248–57. doi: 10.1016/j.humimm.2011.12.015. [DOI] [PubMed] [Google Scholar]
- 69.Mehta RS, Shpall EJ, Rezvani K Cord blood as a source of natural killer cells. Front Med (Lausanne) 2016;2:93. doi: 10.3389/fmed.2015.00093. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 70.Zhao X, Cai L, Hu Y, Wang H Cord-blood natural killer cell-based immunotherapy for cancer. Front Immunol. 2020;11:584099. doi: 10.3389/fimmu.2020.584099. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 71.Alnabhan R, Madrigal A, Saudemont A Differential activation of cord blood and peripheral blood natural killer cells by cytokines. Cytotherapy. 2015;17:73–85. doi: 10.1016/j.jcyt.2014.08.003. [DOI] [PubMed] [Google Scholar]
- 72.Tomchuck SL, Leung WH, Dallas MH Enhanced cytotoxic function of natural killer and CD3+CD56+ cells in cord blood after culture. Biol Blood Marrow Transplant. 2015;21:39–49. doi: 10.1016/j.bbmt.2014.10.014. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 73.Liu E, Ang SOT, Kerbauy L, et al GMP-compliant universal antigen presenting cells (uAPC) promote the metabolic fitness and antitumor activity of armored cord blood CAR-NK cells. Front Immunol. 2021;12:626098. doi: 10.3389/fimmu.2021.626098. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 74.Rezvani K, Rouce RH The application of natural killer cell immunotherapy for the treatment of cancer. Front Immunol. 2015;6:578. doi: 10.3389/fimmu.2015.00578. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 75.Shin HY, Jang S, Woo HJ, et al Cytokine engineered NK-92 therapy to improve persistence and anti-tumor activity. Theranostics. 2023;13:1506–19. doi: 10.7150/thno.79942. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 76.Xiong Q, Zhang H, Ji X, et al A novel membrane-bound interleukin-2 promotes NK-92 cell persistence and anti-tumor activity. Oncoimmunology. 2022;11:2127282. doi: 10.1080/2162402x.2022.2127282. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 77.Sahm C, Schönfeld K, Wels WS Expression of IL-15 in NK cells results in rapid enrichment and selective cytotoxicity of gene-modified effectors that carry a tumor-specific antigen receptor. Cancer Immunol Immunother. 2012;61:1451–61. doi: 10.1007/s00262-012-1212-x. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 78.Chanswangphuwana C, Allan DSJ, Chakraborty M, et al Augmentation of NK cell proliferation and anti-tumor immunity by transgenic expression of receptors for EPO or TPO. Mol Ther. 2021;29:47–59. doi: 10.1016/j.ymthe.2020.09.023. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 79.Mensali N, Dillard P, Fayzullin A, et al “Built-in” PD-1 blocker to rescue NK-92 activity from PD-L1-mediated tumor escape mechanisms. FASEB J. 2021;35:e21750. doi: 10.1096/fj.202100025R. [DOI] [PubMed] [Google Scholar]
- 80.Schmidt P, Raftery MJ, Pecher G Engineering NK cells for CAR therapy-recent advances in gene transfer methodology. Front Immunol. 2021;11:611163. doi: 10.3389/fimmu.2020.611163. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 81.Littwitz E, Francois S, Dittmer U, et al Distinct roles of NK cells in viral immunity during different phases of acute Friend retrovirus infection. Retrovirology. 2013;10:127. doi: 10.1186/1742-4690-10-127. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 82.Goswami R, Subramanian G, Silayeva L, et al Gene therapy leaves a vicious cycle. Front Oncol. 2019;9:297. doi: 10.3389/fonc.2019.00297. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 83.Wu X, Matosevic S Gene-edited and CAR-NK cells: Opportunities and challenges with engineering of NK cells for immunotherapy. Mol Ther Oncolytics. 2022;27:224–38. doi: 10.1016/j.omto.2022.10.011. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 84.Suerth JD, Morgan MA, Kloess S, et al Efficient generation of gene-modified human natural killer cells via alpharetroviral vectors. J Mol Med (Berl) 2016;94:83–93. doi: 10.1007/s00109-015-1327-6. [DOI] [PubMed] [Google Scholar]
- 85.Guven H, Konstantinidis KV, Alici E, et al Efficient gene transfer into primary human natural killer cells by retroviral transduction. Exp Hematol. 2005;33:1320–8. doi: 10.1016/j.exphem.2005.07.006. [DOI] [PubMed] [Google Scholar]
- 86.Nanbakhsh A, Malarkannan S Dextran enhances the lentiviral transduction efficiency of murine and human primary NK cells. Methods Mol Biol. 2020;2097:107–13. doi: 10.1007/978-1-0716-0203-4_7. [DOI] [PubMed] [Google Scholar]
- 87.Allan DSJ, Chakraborty M, Waller GC, et al Systematic improvements in lentiviral transduction of primary human natural killer cells undergoing ex vivo expansion. Mol Ther Methods Clin Dev. 2021;20:559–71. doi: 10.1016/j.omtm.2021.01.008. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 88.Igarashi T, Wynberg J, Srinivasan R, et al Enhanced cytotoxicity of allogeneic NK cells with killer immunoglobulin-like receptor ligand incompatibility against melanoma and renal cell carcinoma cells. Blood. 2004;104:170–7. doi: 10.1182/blood-2003-12-4438. [DOI] [PubMed] [Google Scholar]
- 89.Lin W, Voskens CJ, Zhang X, et al Fc-dependent expression of CD137 on human NK cells: insights into “agonistic” effects of anti-CD137 monoclonal antibodies. Blood. 2008;112:699–707. doi: 10.1182/blood-2007-11-122465. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 90.Denman CJ, Senyukov VV, Somanchi SS, et al Membrane-bound IL-21 promotes sustained ex vivo proliferation of human natural killer cells. PLoS One. 2012;7:e30264. doi: 10.1371/journal.pone.0030264. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 91.Colamartino ABL, Lemieux W, Bifsha P, et al Efficient and robust NK-cell transduction with baboon envelope pseudotyped lentivector. Front Immunol. 2019;10:2873. doi: 10.3389/fimmu.2019.02873. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 92.Nanbakhsh A, Best B, Riese M, et al. Dextran enhances the lentiviral transduction efficiency of murine and human primary NK cells. J Vis Exp 2018:13;55063.
- 93.Pfefferle A, Huntington ND You have got a fast CAR: Chimeric antigen receptor NK cells in cancer therapy. Cancers (Basel) 2020;12:706. doi: 10.3390/cancers12030706. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 94.Robbins GM, Wang M, Pomeroy EJ, et al Nonviral genome engineering of natural killer cells. Stem Cell Res Ther. 2021;12:350. doi: 10.1186/s13287-021-02406-6. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 95.Carlsten M, Levy E, Karambelkar A, et al Efficient mRNA-based genetic engineering of human NK cells with high-affinity CD16 and CCR7 augments rituximab-induced ADCC against lymphoma and targets NK cell migration toward the lymph node-associated chemokine CCL19. Front Immunol. 2016;7:105. doi: 10.3389/fimmu.2016.00105. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 96.Ingegnere T, Mariotti FR, Pelosi A, et al Human CAR NK cells: A new non-viral method allowing high efficient transfection and strong tumor cell killing. Front Immunol. 2019;10:957. doi: 10.3389/fimmu.2019.00957. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 97.Muñoz-López M, García-Pérez JL DNA transposons: nature and applications in genomics. Curr Genomics. 2010;11:115–28. doi: 10.2174/138920210790886871. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 98.Li Y, Hermanson DL, Moriarity BS, et al Human iPSC-derived natural killer cells engineered with chimeric antigen receptors enhance anti-tumor activity. Cell Stem Cell. 2018;23:181–92.e5. doi: 10.1016/j.stem.2018.06.002. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 99.Nguyen DN, Roth TL, Li PJ, et al Polymer-stabilized Cas9 nanoparticles and modified repair templates increase genome editing efficiency. Nat Biotechnol. 2020;38:44–9. doi: 10.1038/s41587-019-0325-6. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 100.Kim KS, Han JH, Park JH, et al Multifunctional nanoparticles for genetic engineering and bioimaging of natural killer (NK) cell therapeutics. Biomaterials. 2019;221:119418. doi: 10.1016/j.biomaterials.2019.119418. [DOI] [PubMed] [Google Scholar]
- 101.Wendel M, Galani IE, Suri-Payer E, Cerwenka A Natural killer cell accumulation in tumors is dependent on IFN-gamma and CXCR3 ligands. Cancer Res. 2008;68:8437–45. doi: 10.1158/0008-5472.Can-08-1440. [DOI] [PubMed] [Google Scholar]
- 102.Chen EB, Zhou ZJ, Xiao K, et al The miR-561-5p/CX3CL1 signaling axis regulates pulmonary metastasis in hepatocellular carcinoma involving CX3CR1+ natural killer cells infiltration. Theranostics. 2019;9:4779–94. doi: 10.7150/thno.32543. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 103.Galland S, Vuille J, Martin P, et al Tumor-derived mesenchymal stem cells use distinct mechanisms to block the activity of natural killer cell subsets. Cell Rep. 2017;20:2891–905. doi: 10.1016/j.celrep.2017.08.089. [DOI] [PubMed] [Google Scholar]
- 104.Terrén I, Orrantia A, Vitallé J, et al NK cell metabolism and tumor microenvironment. Front Immunol. 2019;10:2278. doi: 10.3389/fimmu.2019.02278. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 105.Müller N, Michen S, Tietze S, et al Engineering NK cells modified with an EGFRvIII-specific chimeric antigen receptor to overexpress CXCR4 improves immunotherapy of CXCL12/SDF-1α-secreting glioblastoma. J Immunother. 2015;38:197–210. doi: 10.1097/cji.0000000000000082. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 106.Ng YY, Tay JCK, Wang S CXCR1 expression to improve anti-cancer efficacy of intravenously injected CAR-NK cells in mice with peritoneal xenografts. Mol Ther Oncolytics. 2019;16:75–85. doi: 10.1016/j.omto.2019.12.006. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 107.Tong L, Jiménez-Cortegana C, Tay AHM, et al NK cells and solid tumors: therapeutic potential and persisting obstacles. Mol Cancer. 2022;21:206. doi: 10.1186/s12943-022-01672-z. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 108.Walle T, Kraske JA, Liao B, et al Radiotherapy orchestrates natural killer cell dependent antitumor immune responses through CXCL8. Sci Adv. 2022;8:eabh4050. doi: 10.1126/sciadv.abh4050. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 109.Fitzgerald AA, Wang S, Agarwal V, et al DPP inhibition alters the CXCR3 axis and enhances NK and CD8+ T cell infiltration to improve anti-PD1 efficacy in murine models of pancreatic ductal adenocarcinoma. J Immunother Cancer. 2021;9:e002837. doi: 10.1136/jitc-2021-002837. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 110.Jang ES, Shin JH, Ren G, et al The manipulation of natural killer cells to target tumor sites using magnetic nanoparticles. Biomaterials. 2012;33:5584–92. doi: 10.1016/j.biomaterials.2012.04.041. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 111.Lu C, Guo C, Chen H, et al A novel chimeric PD1-NKG2D-41BB receptor enhances antitumor activity of NK92 cells against human lung cancer H1299 cells by triggering pyroptosis. Mol Immunol. 2020;122:200–6. doi: 10.1016/j.molimm.2020.04.016. [DOI] [PubMed] [Google Scholar]
- 112.Romee R, Schneider SE, Leong JW, et al Cytokine activation induces human memory-like NK cells. Blood. 2012;120:4751–60. doi: 10.1182/blood-2012-04-419283. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 113.Lizana-Vasquez GD, Torres-Lugo M, Dixon RB, et al The application of autologous cancer immunotherapies in the age of memory-NK cells. Front Immunol. 2023;14:1167666. doi: 10.3389/fimmu.2023.1167666. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 114.Chang Y, Jin G, Luo W, et al Engineered human pluripotent stem cell-derived natural killer cells with PD-L1 responsive immunological memory for enhanced immunotherapeutic efficacy. Bioact Mater. 2023;27:168–80. doi: 10.1016/j.bioactmat.2023.03.018. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 115.Töpfer K, Cartellieri M, Michen S, et al DAP12-based activating chimeric antigen receptor for NK cell tumor immunotherapy. J Immunol. 2015;194:3201–12. doi: 10.4049/jimmunol.1400330. [DOI] [PubMed] [Google Scholar]
- 116.Cytlak UM, Dyer DP, Honeychurch J, et al Immunomodulation by radiotherapy in tumour control and normal tissue toxicity. Nat Rev Immunol. 2022;22:124–38. doi: 10.1038/s41577-021-00568-1. [DOI] [PubMed] [Google Scholar]
- 117.Galluzzi L, Humeau J, Buqué A, et al Immunostimulation with chemotherapy in the era of immune checkpoint inhibitors. Nat Rev Clin Oncol. 2020;17:725–41. doi: 10.1038/s41571-020-0413-z. [DOI] [PubMed] [Google Scholar]
- 118.Liu J, Yu Y, Liu C, et al Combinatorial regimens of chemotherapeutic agents: A new perspective on raising the heat of the tumor immune microenvironment. Front Pharmacol. 2022;13:1035954. doi: 10.3389/fphar.2022.1035954. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 119.Klapdor R, Wang S, Morgan MA, et al NK cell-mediated eradication of ovarian cancer cells with a novel chimeric antigen receptor directed against CD44. Biomedicines. 2021;9:1399. doi: 10.3390/biomedicines9101339. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 120.Qian Y, Yang T, Liang H, et al Myeloid checkpoints for cancer immunotherapy. Chin J Cancer Res. 2022;34:460–82. doi: 10.21147/j.issn.1000-9604.2022.05.07. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 121.Liang Y, Liu X, Li K, et al Current situation of programmed cell death protein 1/programmed cell death ligand 1 inhibitors in advanced triple-negative breast cancer. Chin J Cancer Res. 2022;34:117–30. doi: 10.21147/j.issn.1000-9604.2022.02.07. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 122.Ghaedrahmati F, Esmaeil N, Abbaspour M Targeting immune checkpoints: how to use natural killer cells for fighting against solid tumors. Cancer Commun (Lond) 2023;43:177–213. doi: 10.1002/cac2.12394. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 123.Wang F, Wu L, Yin L, et al Combined treatment with anti-PSMA CAR NK-92 cell and anti-PD-L1 monoclonal antibody enhances the antitumour efficacy against castration-resistant prostate cancer. Clin Transl Med. 2022;12:e901. doi: 10.1002/ctm2.901. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 124.André P, Denis C, Soulas C, et al Anti-NKG2A mAb is a checkpoint inhibitor that promotes anti-tumor immunity by unleashing both T and NK cells. Cell. 2018;175:1731–43.e13. doi: 10.1016/j.cell.2018.10.014. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 125.Elia I, Haigis MC Metabolites and the tumour microenvironment: from cellular mechanisms to systemic metabolism. Nat Metab. 2021;3:21–32. doi: 10.1038/s42255-020-00317-z. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 126.Luo W, Qiu J, Zheng L, et al Novel therapies targeting hypoxia mechanism to treat pancreatic cancer. Chin J Cancer Res. 2021;33:216–31. doi: 10.21147/j.issn.1000-9604.2021.02.09. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 127.Wang J, Lupo KB, Chambers AM, et al Purinergic targeting enhances immunotherapy of CD73+ solid tumors with piggyBac-engineered chimeric antigen receptor natural killer cells. J Immunother Cancer. 2018;6:136. doi: 10.1186/s40425-018-0441-8. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 128.Yu L, Liu X, Wang X, et al TIGIT+ TIM-3+ NK cells are correlated with NK cell exhaustion and disease progression in patients with hepatitis B virus-related hepatocellular carcinoma. Oncoimmunology. 2021;10:1942673. doi: 10.1080/2162402x.2021.1942673. [DOI] [PMC free article] [PubMed] [Google Scholar]


