Download as pdf or txt
Download as pdf or txt
You are on page 1of 17

Manufacturing CAR-NK against tumors: Who is the ideal supplier?

Feifei Guo, Yi Zhang, Jiuwei Cui

Citation: Feifei Guo, Yi Zhang, Jiuwei Cui. Manufacturing CAR-NK against tumors: Who is the ideal supplier?. Chin J Cancer
Res 2024; 36(1): 1-16. doi: 10.21147/j.issn.1000-9604.2024.01.01

View online: http://article.cjcrcn.org/article/doi/10.21147/j.issn.1000-9604.2024.01.01

Articles you may be interested in


Immune evasion and therapeutic opportunities based on natural killer cells
Chin J Cancer Res 2023; 35(3): 283 https://doi.org/10.21147/j.issn.1000-9604.2023.03.07

A durable 4-1BB-based CD19 CAR-T cell for treatment of relapsed or refractory non-Hodgkin lymphoma
Chin J Cancer Res 2022; 34(1): 53 https://doi.org/10.21147/j.issn.1000-9604.2022.01.05

Th1 cells inducing IFNγ response improves immunotherapy efficacy in gastric cancer
Chin J Cancer Res 2023; 35(3): 299 https://doi.org/10.21147/j.issn.1000-9604.2023.03.08

Challenges and interventions of chimeric antigen receptor-T cell therapy in solid tumors
Chin J Cancer Res 2023; 35(3): 239 https://doi.org/10.21147/j.issn.1000-9604.2023.03.03

All-trans retinoic acid (ATRA) inhibits insufficient radiofrequency ablation (IRFA)-induced enrichment of tumor-initiating cells in
hepatocellular carcinoma
Chin J Cancer Res 2021; 33(6): 694 https://doi.org/10.21147/j.issn.1000-9604.2021.06.06

Research advances of secretory proteins in malignant tumors


Chin J Cancer Res 2021; 33(1): 115 https://doi.org/10.21147/j.issn.1000-9604.2021.01.12

关注微信公众号,获得更多资讯信息
Review Article

Manufacturing CAR-NK against tumors: Who is the ideal supplier?


Feifei Guo*, Yi Zhang*, Jiuwei Cui

The First Hospital of Jilin University, Cancer Center, Changchun 133021, China
*These authors contributed equally to this work.
Correspondence to: Jiuwei Cui. The First Hospital of Jilin University, Cancer Center, Changchun 133021, China. Email: cuijw@jlu.edu.cn.

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

Submitted Nov 02, 2023. Accepted for publication Jan 12, 2024.
doi: 10.21147/j.issn.1000-9604.2024.01.01
View this article at: https://doi.org/10.21147/j.issn.1000-9604.2024.01.01

Introduction CAR-NK products to help reduce patient waiting times


and treatment costs (5). However, each source has its
Despite the remarkable effectiveness of chimeric antigen
drawbacks. Therefore, in this review, different CAR-NK
receptor-T (CAR-T) cell therapy in human malignancies, it
cell sources are discussed to assist in making an informed
is associated with a plethora of challenges (1). In addition to
choice when applying CAR-NK cells.
optimizing CAR-T cell efficacy, the alternative role of
natural killer (NK) cells has garnered significant interest.
Comparison of CAR-NK cells derived from
The distinct characteristics of chimeric antigen receptor-
different sources
natural killer (CAR-NK) cells highlight their superior
potential for anti-tumor efficacy compared to CAR-T cells Peripheral blood (PB), cord blood (CB), NK cell lines, and
(2). In addition to CAR-mediated cytotoxicity, NK cells
iPSC are the main sources of NK cells for CAR
can lyse cancer cells with downregulated or deficient target
modification (5). NK cells from four promising cell types
antigens via NK-specific cytotoxicity, indicating that CAR-
possess their advantages but also exhibit different problems
NK cells are excellent candidates for eliminating
upon application (Figure 1).
heterogeneous solid tumors (3). CAR-NK therapy is also
believed to be safer than CAR-T cell therapy, given the PB-derived CAR-NK cells
short-term survival of CAR-NK cells in vivo and the
distinctive cytokine spectrum compared to CAR-T cells (4). PB-NK cells
“Multiple sources” is another significant advantage of Human PB mononuclear cells (PBMC) are conventionally
CAR-NK therapy. In addition to autologous peripheral used as NK cell sources in the early adoptive
blood-derived NK cells, multiple allogeneic NK cells immunotherapy (6,7). PB-NK cells have two subsets (8):
further enhance the possibility of fabricating off-the-shelf CD56brightCD16−/dim PB-NK cells that are the mostly

© Chinese Journal of Cancer Research. All rights reserved. www.cjcrcn.org Chin J Cancer Res 2024;36(1):1-16
2 Guo et al. CAR-NK cells: Characteristics and barriers

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.

immature and CD56dimCD16+ that are more mature. Advances in research on PB-CAR-NK cells
CD56brightCD16−/dim PB-NK has longer telomeres (9) PB-CAR-NK cells have demonstrated significant efficacy
and will gradually acquire CD56dim phenotypes (obtaining in treating both hematological and solid malignancies. The
CD16 and KIRs and losing NKG2A) as it matures (10). 2G CAR structure is the most popular. Kruschinski et al.
PB-NK cells used for immunotherapy can be divided into constructed human epidermal growth factor receptor 2
autologous and allogeneic sources. Autologous NK cells (HER2)-specific CAR-NK cells with CD28-CD3ζ
usually exhibit more limited anti-tumor effects than structure. This could override inhibitory signals in primary
allogeneic NK cells (11), since autologous KIR/HLA NK cells and exhibit high-level cytokine release and
matching between NK and tumor cells and degranulation to eradicate tumor cells in HER2-positive
immunosuppressive conditions are critical contributors to carcinoma models (16). Similarly, Imai et al. demonstrated
NK cell inactivation and dysfunction (12). In contrast, that anti-CD19 CAR-NK cells with 4-1BB-CD3ζ structure
mismatched KIR/HLA between allogeneic NK and tumor were capable of surmounting NK resistance and producing
cells excludes interference of inhibitory signals and ensures incremental amounts of INF-γ and GM-CSF to promote
sufficient NK activity (13). However, it should be noted the killing of leukemic cells (17). Recently, additional NK
that T cells in allogeneic peripheral blood must be removed cell-specific CAR structures have been devised to optimize
before NK cell application to avoid graft versus host disease CAR-NK cell functions. For example, a unique CAR-NK
(GVHD) (14). As a popular choice for gene modification, cell line with NKG2D-DAP10-CD3ζ structure designed to
universal accessibility, low cost, and high safety are the exploit NKG2D-MICA/B interactions has exhibited
main advantages of PB-NK. The overexpression of killer remarkable cytotoxicity in acute lymphoblastic leukemia
cell-activating receptors in PB-NK, such as CD16, NKP44, (ALL), osteosarcoma, prostate carcinoma, and
and NKP46, also plays an essential role in enhancing anti- rhabdomyosarcoma models (18).
tumor activity (15). To date, three clinical trials have been conducted to

© Chinese Journal of Cancer Research. All rights reserved. www.cjcrcn.org Chin J Cancer Res 2024;36(1):1-16
Chinese Journal of Cancer Research, Vol 36, No 1 February 2024 3

examine PB-CAR-NK cells. Regarding hematological (NCT05703854 and NCT05922930) (Table 1).
malignancies, one was conducted to explore the efficacy of
haploidentical PB-derived anti-CD19-CAR-NK (4-1BB- NK cell line-derived CAR-NK cells
CD3ζ) in B-lineage ALL patients (NCT00995137), and
NK cell lines
another was carried out to evaluate the anti-tumor effect of
a similar anti-CD19-CAR-NK in patients with refractory In addition to PB/CB-derived NK cells, there are
ALL (NCT01974479). For solid tumors, a phase I clinical numerous mature NK cell lines, such as NK-92, NKL,
trial is being planned by Guangzhou Medical University in HANK1, IMC-1, SNK-6, and SNT-8, all of which can be
China to explore both the efficacy and safety of cultured on a large scale in vitro to provide sufficient cells
autologous/allogeneic broad-spectrum NKG2D-ligand- for application. Among them, NK-92 is the only NK cell
targeting CAR-NK in multiple metastatic solid tumors line approved for clinical trials and is an important supplier
(NCT03415100). Considering the exceptional outcomes for the manufacture of CAR-NK (24).
observed in the corresponding pre-clinical studies, there is The NK-92 cell line has unique molecular
anticipation for noteworthy results from these clinical trials characteristics, such as the expression of CD2+, CD56+,
(Table 1). CD3−, and CD16− (25). NK-92 cells are functionally
characterized by their high activation and dependence on
CB-derived CAR-NK cells IL-2 for their cytotoxic killing capabilities. These cells have
an exceptional ability to recognize tumor cells and exert
CB-NK cells
potent cytotoxic effects (26). NK-92 cells are equipped with
CB-NK cells are NK cells that differentiate from
activating receptors and sufficient cytolytic molecules
hematopoietic stem cells in umbilical CB. CB is an off-the-
(27,28). NK-92 cells express low levels of inhibitory
shelf source of allogeneic NK cells for several reasons.
receptors such as NKG2A to avoid cancer cell-mediated
First, the convenient collection and cryopreservation of CB
inhibition (25). Antibody-dependent cellular cytotoxicity
makes it easy to store (19). Second, compared to other
(ADCC) is another pathway used by NK cells to kill cancer
sources of grafts, CB has fewer T cells, thereby reducing
cells. Despite the low expression of CD16 reported in NK-
the risk of GVHD (20). Moreover, CB provides a more
92 cells, NantKwest successfully engineered an NK cell
stable and higher number of NK cells than PB (21). CB
product with a high affinity for CD16 (29). This
also comprises more NK cell progenitors (22).
modification aimed to address this limitation and enhance
the cytotoxicity of NK-92 cells. Clinical trials registered
Advances in research on CB-CAR-NK
under the identifiers NCT03027128 and NCT03853317
Examination of CB-NK cells has entered clinical trials. A
are currently being conducted to investigate the
phase I/II clinical trial (NCT03056339) was carried out to
test the efficacy and safety of anti-CD19 CAR-CB-NK effectiveness of this approach. For clinical use, the
cells in 11 patients with relapsed or refractory CD19- immortalized NK-92 cell line can be easily cryopreserved
positive cancers. CB-NK cells were designed to co-express in vitro and expanded to large numbers via good
anti-CD19, interleukin (IL)-15, and inducible caspase-9 manufacturing practices (GMP) (30). According to GMP
(iCasp9). IL-15 is a crucial cytokine involved in the standards, Tam et al. successfully developed a highly
proliferation and persistence of NK cells. Suicide genes are efficient protocol that allowed the proliferation of NK-92
safety switches that prevent toxicity. A total of eight (73%) cells more than 200-fold within a 2−2.5-week period. This
patients presented a response, of which seven had complete protocol not only meets clinical immunotherapeutic
remission (CR), and one had remission with respect to requirements but also ensures the quality and safety of
Richter’s transformation with persistent CLL. The most proliferated cells (31).
striking finding of this study was the complete irrelevance
of cytokine release syndrome (CRS)/neurotoxicity/GVHD Advances in research on CAR-NK-92 therapy
and CAR-NK therapy (23). CAR-NK-92 therapy has achieved outstanding results for
Other phase I clinical studies using CB-CAR-NK cells hematological malignancies and solid tumors. Oelsner et al.
with a similar CAR structure (anti-CD19) in patients with constructed a CAR-NK-92 cell line that targeted FMS-like
lymphoma and leukemia have also been launched and are tyrosine kinase 3 (FLT3), which is overexpressed in B-
currently underway. Furthermore, the effects and safety of ALL, and confirmed its notable anti-tumor cytotoxicity in
CB-CAR-NK cells in solid cancers are also being studied SEM B-ALL xenograft models (32). Romanski et al. found

© Chinese Journal of Cancer Research. All rights reserved. www.cjcrcn.org Chin J Cancer Res 2024;36(1):1-16
4 Guo et al. CAR-NK cells: Characteristics and barriers

Table 1 Clinical trials and products of CAR-NK cell therapy

Sources of Product
Trial identifier Tumor type Target Stage Status
NK cells name
PB-NK NCT05645601 JD010 B-cell malignancies CD19 Phase I Recruiting
NCT05410717 − Ovarian cancer, testis cancer Claudin6 Phase I/IIa
and other endometrial cancers
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 Phase I Unknown status
ligands
NCT05020678 NKX019 B-cell malignancies CD19 Phase I Recruiting
NCT04623944 NKX101 AML or MDS NKG2D Phase I Recruiting
ligands
CB-NK NCT05247957 − AML NKG2D Phase I Terminated
ligands
NCT05472558 − B-cell NHL CD19 Phase I Recruiting
NCT04796675 − B-cell malignancies CD19 Phase I Recruiting
NCT05922930 − Ovarian cancer, mesonephric-like TROP2 Phase I/II Not yet recruiting
adenocarcinoma, and pancreatic
cancer
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 CD70 Phase I/II Recruiting
myeloma
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 CD70 Phase I/II Recruiting
and osteosarcoma
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 MUC1 Phase I/II Unknown status
cell lung cancer, pancreatic
carcinoma, triple-negative invasive
breast carcinoma, malignant glioma
of brain, colorectal carcinoma,
gastric carcinoma
NCT03383978 − Glioblastoma HER2 Phase I Recruiting
NCT03656705 − Non-small cell lung cancer −* Phase I Completed
Table 1 (continued)

© Chinese Journal of Cancer Research. All rights reserved. www.cjcrcn.org Chin J Cancer Res 2024;36(1):1-16
Chinese Journal of Cancer Research, Vol 36, No 1 February 2024 5

Table 1 (continued)

Sources of Product
Trial identifier Tumor type Target Stage Status
NK cells name
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 MICA/ Phase I Active, not
cancer, breast cancer, ovarian MICBα3 recruiting
cancer, pancreatic cancer, head and
neck cancer, gastroesophageal
cancer
NCT05336409 CNTY-101 B-cell malignancies CD19 Phase I Recruiting
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.

that engineered NK-92 cells with an anti-CD19 CAR In addition to single CAR modifications, promising
structure could recover their significant cytotoxicity against combination strategies have been used to boost CAR-NK
CD19-expressing B-precursor leukemia cell lines and efficacy in solid tumors. Zhang et al. showed the synergistic
lymphocytes from patients with leukemia (33). A phase I/II effects of regorafenib and CAR-NK-92 cells targeting
clinical trial (NCT02944162) reported the efficacy and epithelial cell adhesion molecules (EpCAM) in colorectal
safety of CAR-NK-92 cells targeting CD33 in three cancer xenograft mouse models (39). A phase I clinical trial
patients with relapsed and refractory acute myeloid (NCT03383978) targeting glioblastoma with anti-HER2
leukemia (AML) (34). Clinical trials NCT02892695 CAR-NK-92 and another phase I clinical trial
exploring the efficacy of 3G CD19-CAR-NK-92 cells and (NCT03656705) designed for non-small cell lung cancer
NCT02742727 exploring the safety of CD7-CAR-NK-92 were initiated to further explore the efficacy and safety of
cells in lymphoma and leukemia are still underway. CAR-NK-92 cells in the treatment of solid clinical tumors
As for solid tumors, CAR-NK-92 also presented (Table 1).
outstanding efficacy. To overcome ovarian cancer, one of
iPSC-derived CAR-NK cells
the most lethal gynecological cancers, Ao et al. designed
anti-folate receptor α (αFR) CAR-NK-92 cells and iPSC-derived NK cells
confirmed their effective killing effect on ovarian cancer The development of iPSCs was first initiated by Takahashi
cells in vitro and ovarian cancer mouse models (35). using Oct3/4, Sox2, c-Myc, and Klf4 (40). Subsequently,
Klapdor et al. constructed a novel dual-CAR structure in human iPSCs were successfully generated and used as
NK-92 cells (anti-CD24 and anti-mesothelin) that showed renewable cell sources (41). Similar to embryonic stem cells
high cytotoxicity in both ovarian cancer cell lines and (ESCs), iPSCs exhibit indefinite proliferation and
primary ovarian cancer cells (36). Han et al. found that pluripotency. Successful induction of iPSCs allows for the
intracranial administration of anti-EGFR CAR-NK-92 convenient acquisition of pluripotent stem cells without
cells inhibited tumor growth and prolonged tumor-bearing embryos. Blood cells derived from iPSCs have
mouse survival in glioblastoma mouse models (37). Esser characteristics similar to those derived from ESCs (42).
et al. showed that CAR-NK-92 cells targeting GD2 showed A robust system for the production of clinical grade
enhanced cytotoxicity in neuroblastoma pre-clinical models iPSC has been developed (43). For iPSC-CAR-NK
(38). In addition, anti-MUC1 CAR-NK-92 cells were also products, CAR structural modifications were executed in
utilized to treat relapsed or refractory solid tumors the undifferentiated state of iPSCs. Subsequently, modified
(NCT02839954). iPSCs are differentiated in a feeder-dependent or feeder-

© Chinese Journal of Cancer Research. All rights reserved. www.cjcrcn.org Chin J Cancer Res 2024;36(1):1-16
6 Guo et al. CAR-NK cells: Characteristics and barriers

free manner in two steps: First, iPSCs are stimulated by Challenges and potential solutions for CAR-NK
stem cell factor (SCF), vascular endothelial growth factor, cells derived from different sources
and bone morphogenic protein 4 to differentiate into
hematopoietic stem cells. Subsequently, CD34+ In vitro expansion and cryopreservation of PB-NK cells
hematopoietic stem cells are selected to further
Due to the low frequency of NK cells in PB, in vitro
differentiate into NK cells under stimulation by IL-3, IL-7,
expansion is necessary to obtain clinical-scale quantities
IL-15, SCF, and FLT3L (44). Finally, differentiated iPSC-
(52). Feeder-dependent and feeder-free approaches are the
CAR-NK cells were co-cultured with artificial antigen-
two main expansion approaches.
presenting cells (aAPC and K562 cells expressing
Co-stimulatory signaling is essential for NK cell
mbIL15/mbIL21 + 41BBL) for clinical-scale expansion
expansion. Feeder cells can provide different co-stimulatory
(45). Zeng et al. established a practical strategy to expand
signals to NK cells via cell-to-cell contact, allowing
the number of NK cells without KIRs, thereby broadening
clinical-scale expansion of NK cells (53). K562, an
the range of eligible patients (46). iPSC-derived NK cells
erythroleukemia cell line lacking HLA antigen expression,
display many of the same characteristics as primary NK
was initially found to be co-stimulatory. Irradiated K562
cells, including the presence of specific markers, such as
cells have been widely used to enhance NK cell expansion
CD56, NKG2D, and CD16 (47). Recent evidence suggests (54). K562, which was modified to express specific co-
that iPSC-derived NK cells have comparable or more stimulatory molecules, exhibited better NK-expansion
potent anti-tumor effects than PB/CB NK cells (48). efficacy in clinical trials. For example, K562-mb15-4-1BBL
Moreover, iPSC-derived NK cells are more suitable for is capable of increasing NK expansion and upregulating
expressing CAR structures than primary NK cells (44). NK activation markers such as CD69, CD25, NKG2D,
NKp30, NKp44, and NKp46. No T cell expansion, genetic
Advances in research on iPSC-CAR-NK cells alterations, or potential oncogenic effects has been
Pre-clinical and clinical studies of iPSC-derived NK cells observed (17,55). MbIL-21 is an excellent substitute for
have been conducted in recent years (49). A high-affinity, mbIL-15. K562 cells expressing mbIL-21 showed enhanced
non-cleavable variant of CD16a was introduced and expansion of NK cells (>47,000-folds in 21 d), and these
engineered in human iPSCs, resulting in the creation of proliferated cells always exhibited increased activation and
potent high-affinity noncleavable variant of CD16a purity without senescence, represented by telomere
(hnCD16)-iPSC-NK cells with improved ADCC shortening (56). The irradiated EBV-transformed
properties (50). Furthermore, iPSC-NK cells armed with lymphoblastoid cell line has also been used to obtain
anti-EpCAM CAR have been developed that show potent clinical-scale NK cells, and these expanded NK cells exhibit
cytotoxicity against EpCAM-positive cancer cells that are superior activation and cytotoxicity compared to naïve NK
resistant to NK cell attack (51). cells (57,58).
Clinical studies have investigated the effectiveness of To further strengthen the safety of feeder-dependent
iPSC-CAR NK cells against various malignancies. The expansion systems, many non-tumor or non-genetically
modified feeder cells have been explored. Two phase I
ELiPSE-1 study (NCT05336409) is currently recruiting
clinical trials examining adoptive NK therapy used
participants to evaluate the safety, pharmacokinetics,
RetroNectin-stimulated T cells (RN-T) to expand
and preliminary efficacy of CNTY-101 in CD19-positive
autologous NK cells, and both obtained sufficient NK cells
B cell malignancies. Furthermore, clinical trials using
with high purity, functionality (high expression of NKG2D
iPSC-CAR-NK cells such as FT576 and FT596 targeting
and CD16), and safety (59,60). Irradiated autologous
BCMA (NCT05182073) and CD19 (NCT04245722 PBMCs are alternate choices (61), and their combination
and NCT04555811), respectively, are underway for with anti-CD16 monoclonal antibodies was confirmed to
examining their therapeutic potential for hematological be an effective approach to expand highly purified cytotoxic
malignancies. In particular, FT536 is specifically designed NK cells with potent anti-tumor function both in vitro and
to target MICA/MICBα3, which are NKG2D ligands in vivo (61,62). Compared to feeder cells, feeder-free
for the treatment of solid cancers (NCT05395052). Due to approaches may facilitate easier licensing as a clinical-grade
the novelty of iPSC-CAR NK cells, more clinical expansion platform, regardless of the infusion of viable
experiments are required to explore their safety and efficacy feeder cells. Based on the existing application of different
(Table 1). non-cell-based activating supplements (e.g., cytokines) for

© Chinese Journal of Cancer Research. All rights reserved. www.cjcrcn.org Chin J Cancer Res 2024;36(1):1-16
Chinese Journal of Cancer Research, Vol 36, No 1 February 2024 7

clinical-grade NK cell expansion, optimization of the cytokines (72). CB-NK cells can also be activated by feeder
combination, adjusting sequencing, and duration of cells. Liu et al. successfully created universal antigen-
exposure time will be more extensively examined in the presenting cells (uAPC) by engineering K562 cells with
future (63,64). increased expression of CD48, 4-1BBL, and mbIL21 to
PB-NK cells exhibit reduced cytotoxicity after promote the expansion and activation of CB-CAR-NK
cryopreservation and long-distance transportation; cells (73).
however, this can be partially overcome by the expansion
process, which largely limits the therapeutic effect of In vivo persistence of NK92 cells
adoptive NK therapy (65). Therefore, the exploration of
Despite the advantages of NK-92 cells over PB/CB-NK
effective cryoprotective agents (CPAs) is a promising
cells in terms of expansion and modification, they still face
strategy. Dimethyl sulfoxide (DMSO), a widely used
challenges due to their limited persistence in vivo. There
cryopreservation reagent, exhibits broad toxicity. It is
are two possible reasons for the limited persistence of NK-
capable of altering the expression of NK cell markers and
92 cells. First, for lymphoma, NK-92 cells must be
impairing NK in vivo functions (66). In view of this, Yao
irradiated before adoptive transfer to eliminate the risk of
et al. designed a biocompatible chitosan-based
tumorigenicity and Epstein-Barr viral susceptibility (74).
nanoparticle, which can mediate efficient DMSO-free NK-
However, irradiation was detrimental to the persistence of
cell cryopreservation via intracellular delivery of non-
NK-92 cells in vivo. Furthermore, the viability and
DMSO CPAs. More importantly, NK cells cryopreserved
proliferation of NK-92 cells are highly dependent on a
in this manner maintain their strong cytotoxicity,
continuous supply of low-dose IL-2. In the absence of IL-
degranulation, and cytokine production functions (67).
2, NK-92 cells exhibited a rapid decline in survival, leading
Simultaneous maturation and expansion of CB-NK cells to death in 72 h. Therefore, when introduced into the
human body, the absence of IL-2 poses a significant threat
The use of CB-NK cells presents two significant challenges to the longevity of NK-92 cells (25).
for investigators: the limited number of NK cells in a CB Enhancing the persistence of NK-92 cells can be
unit and their naïve phenotype. Although the percentage of achieved via gene modifications, particularly by targeting
NK cells was similar or even higher in CB than in PB, the various elements, such as cytokines. For example, NK-92
total number of NK cells was limited by the small volume cells can be modified by attaching IL-2 to their plasma
of a single CB unit (68). In addition, CB-NK cells are less membranes to stimulate nearby IL-2 receptors. This
mature than their PB counterparts (69). CB-NK cells membrane-bound protein (MBP) NK-92 cells exhibited
exhibit a different phenotype characterized by decreased exogenous IL-2 independent proliferation and showed
expression of molecules such as CD16 and granzyme B and improved infiltration and persistent activity in an A549
increased expression of NKG2A and CXCR3 (68,70). The spheroid solid tumor model (75). Similarly, mbIL-2-
immature phenotype observed in umbilical CB-NK cells is expressing NK-92 cells showed improved persistence and
indicative of reduced cytotoxicity and increased homing enhanced anti-tumor activity in a leukemia xenograft model
propensity. Consequently, CB-NK cells generally (76). IL-15 is another cytokine commonly used in
demonstrate lower anti-cancer activity than PB-NK cells. laboratory and clinical investigations to sustain survival and
Therefore, any attempt to expand CB-NK cells must be enhance the cytotoxicity of NK cells. NK-92 cells co-
accompanied by an effort to enhance their activation. expressing IL-15 and EpCAM/breast cancer-specific CAR
The simultaneous resolution of insufficient in vitro showed persistent and increased cytotoxicity against
expansion and immature killing capacity may be achieved EpCAM-expressing breast carcinoma cells in vitro (77).
via the use of expansion techniques, which commonly In addition to cytokines, other elements can be modified
exploit the benefits of interleukins, particularly IL-2 and to optimize the persistence of NK-92 cells. For example, in
IL-15 (71). Through ex vivo expansion technology, the vivo studies have shown that NK-92 cells expressing
number of NK cells can be substantially augmented, with erythropoietin or thrombopoietin receptors have a longer
an approximately (1,800−2,400)-fold increase observed in lifespan (78). Similarly, NK-92 cells with truncated
fresh or cryopreserved CB units (53). Furthermore, the PD-1 (tPD-1) exhibit enhanced persistence compared to
activation and maturation of NK cells, contributing to their standard NK-92 cells, as tPD-1 blocks the PD-1/PD-L1
effective killing abilities, are achieved via the influence of inhibition (79).

© Chinese Journal of Cancer Research. All rights reserved. www.cjcrcn.org Chin J Cancer Res 2024;36(1):1-16
8 Guo et al. CAR-NK cells: Characteristics and barriers

Common challenges associated with CAR-NK cells investigated the delicate balance between gene editing
efficiency and expansion potential and showed that the
Gene modification of CAR-NK products ideal pre-activation duration of feeder cells is 5−7 d prior to
Overcoming the resistance of NK cells to transduction is a the lentiviral transduction (87). EBV-transformed
significant hurdle in the successful and efficient gene lymphoblastoid cell lines and genetically modified K562
modification of CAR-NK cells. Compared to T cells, NK cells are feeder cells that are commonly used to achieve
cells show a higher sensitivity to foreign genetic materials
clinical-scale NK cell proliferation (23,88). Mechanically,
and a lower transduction efficiency (80). Mechanically,
the secretion of IL-2 by feeder cells serves as a highly
pattern recognition receptors in NK cells recognize foreign
effective enhancer of NK cell expansion and transduction
genetic materials and trigger an innate defense against
(57). Furthermore, physical contact between feeder cells
them (81). Therefore, the potential risks of insertional gene
and NK cells, as well as the binding of ligands on the feeder
mutations should not be overlooked. In the presence of
cells (e.g., 4-1BBL (CD137L), mbIL-15, and mbIL-21) to
high titers of vital vectors, NK cells encounter the potential
their corresponding stimulatory receptors on NK cells, is
genotoxicity associated with insertional mutations (82).
crucial for successful expansion of NK cells (89,90).
Currently, transfection methods are divided into two
types: viral and non-viral. Viral vectors have emerged as (3) Vector and transduction enhancer
highly efficient and superior gene modification tools The design of new vectors appears to be another feasible
compared to non-viral alternatives (83). As an important method. One such example is the use of baboon envelope-
component of viral transfection systems, lentiviral pseudotyped lentiviral vectors, which have been developed
to promote viral entry. Colamartino et al. demonstrated
transfection has been used in phase I/II clinical research on
that these vectors achieved a transduction efficiency of
CAR NK cell therapy for hematological (NCT05472558)
38.3%±23.8% in NK cells, which increased to 58.4%±
and solid (NCT05410717) malignancies. Other retroviral
7.8% after re-expansion (91).
vectors are also primary choices to modify NK cell genes.
The use of transduction enhancers, such as dextran and
For example, Suerth et al. confirmed that alpha-retroviral
IL-2, is another option. Müller et al. conducted a
vectors are useful tools for the application of NK cell-
comparative study of two different retroviral vector
mediated cancer immunotherapy (84).
platforms with the addition of retronectin and vectofusin-1.
To date, many efforts have been made to enhance the
They found that the combination of RD114-TR
transduction efficiency. These include the implementation
pseudotyped retroviral and vectofusin-1 was the optimal
of multiple rounds of transduction, co-culture techniques
strategy to engineer PB-derived NK cells (92).
involving feeder cells, and the development and refinement
(4) Non-viral gene modification
of novel vector designs. Considering the restricted vector capacity and potential
(1) Multiple rounds of transduction risk of tumorigenicity associated with viral transfection,
Adopting multiple rounds of transduction represents a various non-viral vectors have been extensively investigated
direct approach to improving the efficiency of gene in recent years to overcome these limitations.
expression. Guven et al. observed that the percentage of Electroporation, nucleofection, and lipofection are the
NK cells transduced on d 21 was higher in cells that most commonly used non-viral transduction methods (93).
underwent two rounds of transduction than in cells that Compared to lipofection, electroporation has shown
underwent a single round of transduction (75.4% vs. greater efficiency and suitability for clinical translation,
51.9%) (85). In addition, in the presence of the lentiviral although it may be less cost-effective (94). mRNAs are the
transduction enhancer dextran, the transduction rate of NK main cargo involved in non-viral transduction. Carlsten
cells reached 40% after one round of transduction and et al. used electroporation to deliver the mRNA of high-
increased to 100% after two rounds (86). Therefore, affinity CD16 and the chemokine receptor C-C motif
repeated transduction may be a potential method to chemokine receptor 7 to primary NK cells. This resulted in
increase transduction efficiency. 95% of NK cells successfully expressing the target mRNA.
(2) Co-culture with feeder cells The engineered cells exhibit enhanced migration and
Feeder cells can be used to improve the production of cytotoxicity against antibody-coated lymphoma cells (95).
CAR-NK cells. Despite their potential impact on viral However, while mRNA has a lower risk of insertional
transfection efficiency, feeder cells effectively enhance the mutation as it cannot integrate into DNA, it poses a
recovery and expansion of transfected NK cells. Allan et al. limitation for non-viral transduction methods in achieving

© Chinese Journal of Cancer Research. All rights reserved. www.cjcrcn.org Chin J Cancer Res 2024;36(1):1-16
Chinese Journal of Cancer Research, Vol 36, No 1 February 2024 9

permanent CAR structure expression. Additionally, the Second, certain anti-tumor agents, such as radiotherapy
limited capacity of mRNA as a cargo hinders the and autophagy inhibitors, can disrupt normal tumor
development of non-viral transduction (96). processes, leading to increased secretion of chemokine
Various strategies have been explored to overcome the ligands (107,108). For example, dipeptidyl peptidase
aforementioned hurdles, such as the utilization of DNA inhibitors and curaxins have been shown to enhance the
transposons, CRISPR/Cas9 technology, and nanoparticles. secretion of CXCL9 and CXCL10 in different solid
DNA transposons have become popular non-viral gene tumors, promoting the infiltration of CXCR3+ NK cells
delivery tools for achieving stable gene expression (97). into the TME (109). In addition, nanoparticles can
Transposon-mediated CAR transduction, especially with facilitate NK cell infiltration. For instance, magnetic
piggyBac and sleeping beauty transposon systems, has nanoparticles loaded with NK-92 cells showed a 17-fold
shown promising efficacy in achieving stable CAR increase in infiltration when a magnetic field was applied
expression in NK cells through a close-to-random profile compared to normal NK-92 cells (110).
of genomic integration (98). Another promising approach is (2) Optimization of CAR-NK cells
the combination of non-viral gene delivery methods with To improve the function of NK cells, it is important to
CRISPR delivery systems (99). Specifically, Nguyen et al. directly modify NK cells to overcome unfavorable
electroporated Cas9 ribonucleoproteins in NK cells, which conditions in the TME. One promising approach is to
improved the transfection efficiency. Moreover, adapt and modulate CAR-NK cells to respond effectively
multifunctional nanoparticles (MF-NPs) can be used to to the components of TME. Therefore, reengineering the
genetically express anti-EGFR-CAR structures on NK cells CAR structure has shown great potential. A unique CAR,
and image MF-NP-loaded NK cells in vivo (100). namely the novel chimeric costimulatory converting
receptor (CCCR), was designed to address this challenge.
Challenges of CAR-NK cells in solid tumor treatment CCCR incorporates the domain of PD1 (extracellular),
CAR-NK cells from different sources have limited NKG2D (transmembrane and cytoplasmic), and 41BB
effectiveness in solid tumor treatment and yield (cytoplasmic). This design aimed to convert the inhibitory
unsatisfactory outcomes. This can be attributed to several PD-1 signal into an activating one (111). When expressed
factors. First, decreased expression of chemokine ligands, in NK-92 cells, CCCR-NK-92 cells were able to overcome
such as CXCL1 and CXCL9, impairs NK cell trafficking immunosuppressive TME and exhibited enhanced anti-
ability, restricting their infiltration into tumor sites (101). tumor efficacy.
For example, in hepatocellular carcinoma, miR-561-5p Memory-like NK cells have increased production of
overexpression downregulates CXCL1 expression and IFN-γ, which is related to the upregulation of killing-
blocks CXCR1+NK cell infiltration (102). Furthermore, activating receptors and downregulation of killing
the immunosuppressive characteristics and the abnormal inhibitory receptors. This ultimately leads to a superior
metabolic profile of tumor microenvironment (TME) killing effect compared to common NK cells (112).
hinder the survival and function of NK cells (103,104). Inducing immunological memory of NK cells is an
Fortunately, various strategies have shown promise in alternative method to enhance NK cell activity in solid
addressing these challenges in pre-clinical tumor models. tumors (113). For example, Chang et al. developed PD-L1-
(1) Enhancing infiltration of CAR-NK cells induced memory-like iPSC-NK cells that respond to PD-
One approach to enhance the infiltration of NK cells into L1 signaling, resulting in enhanced proliferation and
tumor sites is to increase the expression of chemokines in cytotoxicity. In a mouse xenograft model, these cells
NK cells and stimulate cancer cells to secrete the exhibited higher anti-tumor cytotoxicity than other CAR-
corresponding chemokine ligands. This can be achieved by NK cells (114).
various methods. First, NK cells can be designed to co- Additionally, the effectiveness of NK cells with different
express a CAR structure and chemokine receptors, such as CAR structures may vary depending on the type of cancer.
CXCR3 and CXCR4, which are key mediators of NK cell For example, in prostate cancer mouse models, anti-PSCA-
trafficking. Müller et al. successfully enhanced tumor DAP12 CAR-NK cells showed greater cytotoxicity
infiltration by NK cells by designing novel NK cells with compared to anti-PSCA-CD3ζ CAR-NK cells (115).
EGFRvIII-specific CAR and CXCR4 (105). Similarly, Similarly, Li et al. evaluated different CAR structures in
CXCR1-engineered NK cells exhibited increased iPSC-NK cells and found that NKG2D-2B4-CD3ζ
infiltration of human tumors in a xenograft model (106). CAR exhibited the strongest activating effect on NK

© Chinese Journal of Cancer Research. All rights reserved. www.cjcrcn.org Chin J Cancer Res 2024;36(1):1-16
10 Guo et al. CAR-NK cells: Characteristics and barriers

cells in an ovarian cancer model (98). Therefore, it is have been developed to block these inhibitory checkpoints
crucial to compare and optimize various CARs for different and restore CAR-NK cell function to overcome immune
types of cancer to maximize the cytotoxic effects of CAR- inhibition (122). Wang et al. showed that combining anti-
NK cells. PSMA CAR NK-92 cells with an anti-PD-L1 antibody
(3) TME modification improves anti-tumor efficacy in a castration-resistant
In addition, it is important to regulate the immune and prostate cancer mouse model (123). Furthermore, clinical
metabolic conditions in the TME to enhance the trials have confirmed that monalizumab, an anti-NKG2A
effectiveness of CAR-NK cells. Various agents, such as monoclonal antibody, improves NK cell activity in solid
chemotherapy and immune checkpoint inhibitors (ICIs), cancers (124). The combination of monalizumab with
can modulate the immune response in the TME and CAR-NK cell therapy has the potential to enhance its
improve the persistence and cytotoxicity of NK cells therapeutic effects.
(116,117). Chemotherapy can directly activate NK cells Additionally, abnormal metabolic conditions in TME,
and modify immune response in the TME to support NK such as hypoxia, nutrient deprivation, and accumulation of
cell functions. It achieves this by improving the metabolites (e.g., reactive oxygen species and adenosine),
immunogenicity of cancer cells and regulating the have a significant effect on the metabolism and energy
recruitment and function of immunosuppressive cells (118). supply of NK cells, ultimately affecting their anti-tumor
For example, Klapdor et al. found that treating CD44- properties (125,126). Therefore, modulating metabolism
positive ovarian cancer cells with anti-CD44 NK cells and can be a promising method to enhance the performance of
cisplatin simultaneously improved the anti-tumor capacity NK cells in TME. For example, combining NKG2D-
of NK cells (119). specific CAR-NK-92 cells with CD73 blockade improves
ICIs are effective in regulating immune interactions therapeutic efficacy in CD73+ human lung cancer models
between NK and tumor cells. In solid cancers, prominent (127). CD73 is associated with the accumulation of
inhibitory checkpoints, such as PD-1, NKG2A, and KIRs, adenosine, a metabolite with immunosuppressive effects
can inhibit NK cell activation (120,121). Various antibodies 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.

© Chinese Journal of Cancer Research. All rights reserved. www.cjcrcn.org Chin J Cancer Res 2024;36(1):1-16
Chinese Journal of Cancer Research, Vol 36, No 1 February 2024 11

Conclusions and perspective 6. Ettinghausen SE, Moore JG, White DE, et al.
Hematologic effects of immunotherapy with
CAR-NK cells derived from PB, CB, and NK-92 cell lines lymphokine-activated killer cells and recombinant
and iPSCs are effective in anti-tumor therapy. The interleukin-2 in cancer patients. Blood 1987;69:
abundant allogeneic suppliers of NK cells undoubtedly 1654-60.
provide a promising opportunity for clinical-scale 7. Liu X, Ran R, Shao B, et al. Combined peripheral
manufacturing and application of CAR-NK therapy. natural killer cell and circulating tumor cell
However, each source also has its problems that need to be enumeration enhance prognostic efficiency in
solved and is associated with common challenges that patients with metastatic triple-negative breast cancer.
impede effective CAR-NK production. Although CAR-NK Chin J Cancer Res 2018;30:315-26.
therapy has shown outstanding anti-tumor effects on pre- 8. Cooper MA, Fehniger TA, Caligiuri MA. The
clinical studies of both hematological and solid tumors, biology of human natural killer-cell subsets. Trends
corresponding clinical trials with CAR-NK cells are just Immunol 2001;22:633-40.
beginning, and many critical results have not yet been 9. Romagnani C, Juelke K, Falco M, et al.
published. Therefore, to obtain reliable clinical-level off- CD56brightCD16- killer Ig-like receptor- NK cells
the-shelf CAR-NK products, follow-up comparisons and display longer telomeres and acquire features of
verification of the clinical efficacy and safety of CAR-NK CD56dim NK cells upon activation. J Immunol
cells from different sources and optimization and 2007;178:4947-55.
standardization of CAR-NK manufacturing processes are 10. Domaica CI, Fuertes MB, Uriarte I, et al. Human
required. natural killer cell maturation defect supports in vivo
CD56(bright) to CD56(dim) lineage development.
Acknowledgements PloS One 2012;7:e51677.
11. Parkhurst MR, Riley JP, Dudley ME, et al. Adoptive
None.
transfer of autologous natural killer cells leads to
high levels of circulating natural killer cells but does
Footnote
not mediate tumor regression. Clin Cancer Res 2011;
Conflicts of Interest: These authors have no conflicts of 17:6287-97.
interest to declare. 12. Carrega P, Pezzino G, Queirolo P, et al.
Susceptibility of human melanoma cells to
References autologous natural killer (NK) cell killing: HLA-
related effector mechanisms and role of unlicensed
1. June CH, O’Connor RS, Kawalekar OU, et al. CAR NK cells. PLoS One 2009;4:e8132.
T cell immunotherapy for human cancer. Science 13. Morvan M, David G, Sébille V, et al. Autologous
2018;359:1361-5. and allogeneic HLA KIR ligand environments and
2. Xie G, Dong H, Liang Y, et al. CAR-NK cells: A activating KIR control KIR NK-cell functions. Eur J
promising cellular immunotherapy for cancer. Immunol 2008;38:3474-86.
EBioMedicine 2020;59:102975. 14. Koepsell SA, Miller JS, McKenna DH Jr. Natural
3. Peng X, Chen L, Chen L, et al. Chimeric antigen killer cells: a review of manufacturing and clinical
receptor-natural killer cells: Novel insight into utility. Transfusion 2013;53:404-10.
immunotherapy for solid tumors (Review). Exp Ther 15. Borah A, Zhang J, Guo F, et al. Immune evasion and
Med 2021;21:340. therapeutic opportunities based on natural killer
4. Karadimitris A. Cord blood CAR-NK cells: cells. Chin J Cancer Res 2023;35:283-98.
Favorable initial efficacy and toxicity but durability 16. Kruschinski A, Moosmann A, Poschke I, et al.
of clinical responses not yet clear. Cancer cell 2020; Engineering antigen-specific primary human NK
37:426-7. cells against HER-2 positive carcinomas. Proc Natl
5. Siegler EL, Zhu Y, Wang P, et al. Off-the-shelf Acad Sci USA 2008;105:17481-6.
CAR-NK cells for cancer immunotherapy. Cell 17. Imai C, Iwamoto S, Campana D. Genetic
Stem Cell 2018;23:160-1. modification of primary natural killer cells

© Chinese Journal of Cancer Research. All rights reserved. www.cjcrcn.org Chin J Cancer Res 2024;36(1):1-16
12 Guo et al. CAR-NK cells: Characteristics and barriers

overcomes inhibitory signals and induces specific its impact on natural killer cell research and
killing of leukemic cells. Blood 2005;106:376-83. treatment of cancer. Cytotherapy 2023;25:451-7.
18. Chang YH, Connolly J, Shimasaki N, et al. A 30. Pasley S, Zylberberg C, Matosevic S. Natural killer-
chimeric receptor with NKG2D specificity enhances 92 cells maintain cytotoxic activity after long-term
natural killer cell activation and killing of tumor cryopreservation in novel DMSO-free media.
cells. Cancer Res 2013;73:1777-86. Immunol Lett 2017;192:35-41.
19. Munoz J, Shah N, Rezvani K, et al. Concise review: 31. Tam YK, Martinson JA, Doligosa K, et al. Ex vivo
umbilical cord blood transplantation: past, present, expansion of the highly cytotoxic human natural
and future. Stem Cells Transl Med 2014;3:1435-43. killer-92 cell-line under current good manufacturing
20. Knorr DA, Bachanova V, Verneris MR, et al. practice conditions for clinical adoptive cellular
Clinical utility of natural killer cells in cancer immunotherapy. Cytotherapy 2003;5:259-72.
therapy and transplantation. Semin Immunol 2014; 32. Oelsner S, Waldmann A, Billmeier A, et al.
26:161-72. Genetically engineered CAR NK cells display
21. Lin SJ, Kuo ML. Cytotoxic function of umbilical selective cytotoxicity against FLT3-positive B-ALL
cord blood natural killer cells: relevance to adoptive and inhibit in vivo leukemia growth. Int J Cancer
immunotherapy. Pediatr Hematol Oncol 2011;28: 2019;145:1935-45.
640-6. 33. Romanski A, Uherek C, Bug G, et al. CD19-CAR
22. Rutella S, Bonanno G, Marone M, et al. engineered NK-92 cells are sufficient to overcome
Identification of a novel subpopulation of human NK cell resistance in B-cell malignancies. J Cell Mol
cord blood CD34-CD133-CD7-CD45+lineage- Med 2016;20:1287-94.
cells capable of lymphoid/NK cell differentiation 34. Tang X, Yang L, Li Z, et al. First-in-man clinical
after in vitro exposure to IL-15. J Immunol 2003;171: trial of CAR NK-92 cells: safety test of CD33-CAR
2977-88. NK-92 cells in patients with relapsed and refractory
23. Liu E, Marin D, Banerjee P, et al. Use of CAR- acute myeloid leukemia. Am J Cancer Res 2018;8:
transduced natural killer cells in CD19-positive 1083-9.
lymphoid tumors. N Engl J Med 2020;382:545-53. 35. Ao X, Yang Y, Li W, et al. Anti-αFR CAR-
24. Zhang J, Zheng H, Diao Y. Natural killer cells and engineered NK-92 cells display potent cytotoxicity
current applications of chimeric antigen receptor- against αFR-positive ovarian cancer. J Immunother
modified NK-92 cells in tumor immunotherapy. Int 2019;42:284-96.
J Mol Sci 2019;20:317. 36. Klapdor R, Wang S, Morgan M, et al.
25. Gong J, Maki G, Klingemann HG. Characterization Characterization of a novel third-generation anti-
of a human cell line (NK-92) with phenotypical and CD24-CAR against ovarian cancer. Int J Mol Sci
functional characteristics of activated natural killer 2019;20:660.
cells. Leukemia 1994;8:652-8. 37. Han J, Chu J, Keung Chan W, et al. CAR-
26. Klingemann H, Boissel L, Toneguzzo F. Natural engineered NK cells targeting wild-type EGFR and
killer cells for immunotherapy — Advantages of the EGFRvIII enhance killing of glioblastoma and
NK-92 cell line over blood NK cells. Front patient-derived glioblastoma stem cells. Sci Rep
Immunol 2016;7:91. 2015;5:11483.
27. Boissel L, Betancur-Boissel M, Lu W, et al. 38. Esser R, Müller T, Stefes D, et al. NK cells
Retargeting NK-92 cells by means of CD19- and engineered to express a GD2-specific antigen
CD20-specific chimeric antigen receptors compares receptor display built-in ADCC-like activity against
favorably with antibody-dependent cellular tumour cells of neuroectodermal origin. J Cell Mol
cytotoxicity. Oncoimmunology 2013;2:e26527. Med 2012;16:569-81.
28. Maki G, Klingemann HG, Martinson JA, et al. 39. Zhang Q, Zhang H, Ding J, et al. Combination
Factors regulating the cytotoxic activity of the therapy with EpCAM-CAR-NK-92 cells and
human natural killer cell line, NK-92. J Hematother regorafenib against human colorectal cancer models.
Stem Cell Res 2001;10:369-83. J Immunol Res 2018;2018:4263520.
29. Klingemann H. The NK-92 cell line-30 years later: 40. Takahashi K, Yamanaka S. Induction of pluripotent

© Chinese Journal of Cancer Research. All rights reserved. www.cjcrcn.org Chin J Cancer Res 2024;36(1):1-16
Chinese Journal of Cancer Research, Vol 36, No 1 February 2024 13

stem cells from mouse embryonic and adult 53. Kundu S, Gurney M, O’Dwyer M. Generating
fibroblast cultures by defined factors. Cell 2006;126: natural killer cells for adoptive transfer: expanding
663-76. horizons. Cytotherapy 2021;23:559-66.
41. Takahashi K, Tanabe K, Ohnuki M, et al. Induction 54. Robertson MJ, Cameron C, Lazo S, et al.
of pluripotent stem cells from adult human Costimulation of human natural killer cell
fibroblasts by defined factors. Cell 2007;131:861-72. proliferation: role of accessory cytokines and cell
42. Choi KD, Yu J, Smuga-Otto K, et al. Hematopoietic contact-dependent signals. Nat Immun 1996;15:
and endothelial differentiation of human induced 213-26.
pluripotent stem cells. Stem Cells 2009;27:559-67. 55. Fernández L, Leivas A, Valentín J, et al. How do we
43. Valamehr B, Robinson M, Abujarour R, et al. manufacture clinical-grade interleukin-15-stimulated
Platform for induction and maintenance of natural killer cell products for cancer treatment.
transgene-free hiPSCs resembling ground state Transfusion 2018;58:1340-7.
pluripotent stem cells. Stem Cell Reports 2014;2: 56. Somanchi SS, Lee DA. Ex vivo expansion of human
366-81. NK cells using K562 engineered to express
44. Knorr DA, Ni Z, Hermanson D, et al. Clinical-scale membrane bound IL21. Methods Mol Biol 2016;
derivation of natural killer cells from human 1441:175-93.
pluripotent stem cells for cancer therapy. Stem Cells 57. Granzin M, Stojanovic A, Miller M, et al. Highly
Transl Med 2013;2:274-83. efficient IL-21 and feeder cell-driven ex vivo
45. Zhu H, Kaufman DS. An improved method to expansion of human NK cells with therapeutic
produce clinical-scale natural killer cells from human activity in a xenograft mouse model of melanoma.
pluripotent stem cells. Methods Mol Biol 2019;2048: Oncoimmunology 2016;5:e1219007.
107-19. 58. Granzin M, Soltenborn S, Müller S, et al. Fully
46. Zeng J, Tang SY, Toh LL, et al. Generation of “Off- automated expansion and activation of clinical-grade
the-Shelf” natural killer cells from peripheral blood natural killer cells for adoptive immunotherapy.
cell-derived induced pluripotent stem cells. Stem Cytotherapy 2015;17:621-32.
Cell Reports 2017;9:1796-812. 59. Sakamoto N, Ishikawa T, Kokura S, et al. Phase I
47. Goldenson BH, Hor P, Kaufman DS. iPSC-derived clinical trial of autologous NK cell therapy using
natural killer cell therapies — expansion and novel expansion method in patients with advanced
targeting. Front Immunol 2022;13:841107. digestive cancer. J Trans Med 2015;13:277.
48. Hermanson DL, Bendzick L, Pribyl L, et al. Induced 60. Ishikawa T, Okayama T, Sakamoto N, et al. Phase I
pluripotent stem cell-derived natural killer cells for clinical trial of adoptive transfer of expanded natural
treatment of ovarian cancer. Stem Cells 2016;34: killer cells in combination with IgG1 antibody in
93-101. patients with gastric or colorectal cancer. Int J
49. Saetersmoen ML, Hammer Q, Valamehr B, et al. Cancer 2018;142:2599-609.
Off-the-shelf cell therapy with induced pluripotent 61. Parkhurst MR, Riley JP, Dudley ME, et al. Adoptive
stem cell-derived natural killer cells. Semin transfer of autologous natural killer cells leads to
Immnopathol 2019;41:59-68. high levels of circulating natural killer cells but does
50. Zhu H, Blum RH, Bjordahl R, et al. Pluripotent not mediate tumor regression. Clin Cancer Res
stem cell-derived NK cells with high-affinity 2011;17:6287-97.
noncleavable CD16a mediate improved antitumor 62. Lee HR, Son CH, Koh EK, et al. Expansion of
activity. Blood 2020;135:399-410. cytotoxic natural killer cells using irradiated
51. Tang SY, Zha S, Du Z, et al. Targeted integration of autologous peripheral blood mononuclear cells and
EpCAM-specific CAR in human induced pluripotent anti-CD16 antibody. Sci Rep 2017;7:11075.
stem cells and their differentiation into NK cells. 63. Alici E, Sutlu T, Björkstrand B, et al. Autologous
Stem Cell Res Ther 2021;12:580. antitumor activity by NK cells expanded from
52. Fang F, Wang W, Chen M, et al. Technical myeloma patients using GMP-compliant
advances in NK cell-based cellular immunotherapy. components. Blood 2008;111:3155-62.
Cancer Bio Med 2019;16:647-54. 64. Johnson C, Jesuraj NJ. Seeding density is critical for

© Chinese Journal of Cancer Research. All rights reserved. www.cjcrcn.org Chin J Cancer Res 2024;36(1):1-16
14 Guo et al. CAR-NK cells: Characteristics and barriers

optimizing scalable feeder-free natural killer cell 15 in NK cells results in rapid enrichment and
expansion. Cytotherapy 2020;22:S126. selective cytotoxicity of gene-modified effectors that
65. Damodharan SN, Walker KL, Forsberg MH, et al. carry a tumor-specific antigen receptor. Cancer
Analysis of ex vivo expanded and activated clinical- Immunol Immunother 2012;61:1451-61.
grade human NK cells after cryopreservation. 78. Chanswangphuwana C, Allan DSJ, Chakraborty M,
Cytotherapy 2020;22:450-7. et al. Augmentation of NK cell proliferation and
66. Yao X, Matosevic S. Cryopreservation of NK and T anti-tumor immunity by transgenic expression of
cells without DMSO for adoptive cell-based receptors for EPO or TPO. Mol Ther 2021;29:
immunotherapy. BioDrugs 2021;35:529-45. 47-59.
67. Yao X, Jovevski JJ, Todd MF, et al. Nanoparticle- 79. Mensali N, Dillard P, Fayzullin A, et al. “Built-in”
mediated intracellular protection of natural killer PD-1 blocker to rescue NK-92 activity from PD-L1-
cells avoids cryoinjury and retains potent antitumor mediated tumor escape mechanisms. FASEB J
functions. Adv Sci (Weinh) 2020;7:1902938. 2021;35:e21750.
68. Luevano M, Daryouzeh M, Alnabhan R, et al. The 80. Schmidt P, Raftery MJ, Pecher G. Engineering NK
unique profile of cord blood natural killer cells cells for CAR therapy-recent advances in gene
balances incomplete maturation and effective killing transfer methodology. Front Immunol 2021;11:
function upon activation. Hum Immunol 2012;73: 611163.
248-57. 81. Littwitz E, Francois S, Dittmer U, et al. Distinct
69. Mehta RS, Shpall EJ, Rezvani K. Cord blood as a roles of NK cells in viral immunity during different
source of natural killer cells. Front Med (Lausanne) phases of acute Friend retrovirus infection.
2016;2:93. Retrovirology 2013;10:127.
70. Zhao X, Cai L, Hu Y, Wang H. Cord-blood natural 82. Goswami R, Subramanian G, Silayeva L, et al. Gene
killer cell-based immunotherapy for cancer. Front therapy leaves a vicious cycle. Front Oncol 2019;9:
Immunol 2020;11:584099. 297.
71. Alnabhan R, Madrigal A, Saudemont A. Differential 83. Wu X, Matosevic S. Gene-edited and CAR-NK
activation of cord blood and peripheral blood natural cells: Opportunities and challenges with engineering
killer cells by cytokines. Cytotherapy 2015;17:73-85. of NK cells for immunotherapy. Mol Ther
72. Tomchuck SL, Leung WH, Dallas MH. Enhanced Oncolytics 2022;27:224-38.
cytotoxic function of natural killer and CD3+CD56+ 84. Suerth JD, Morgan MA, Kloess S, et al. Efficient
cells in cord blood after culture. Biol Blood Marrow generation of gene-modified human natural killer
Transplant 2015;21:39-49. cells via alpharetroviral vectors. J Mol Med (Berl)
73. Liu E, Ang SOT, Kerbauy L, et al. GMP-compliant 2016;94:83-93.
universal antigen presenting cells (uAPC) promote 85. Guven H, Konstantinidis KV, Alici E, et al. Efficient
the metabolic fitness and antitumor activity of gene transfer into primary human natural killer cells
armored cord blood CAR-NK cells. Front Immunol by retroviral transduction. Exp Hematol 2005;33:
2021;12:626098. 1320-8.
74. Rezvani K, Rouce RH. The application of natural 86. Nanbakhsh A, Malarkannan S. Dextran enhances the
killer cell immunotherapy for the treatment of lentiviral transduction efficiency of murine and
cancer. Front Immunol 2015;6:578. human primary NK cells. Methods Mol Biol 2020;
75. Shin HY, Jang S, Woo HJ, et al. Cytokine 2097:107-13.
engineered NK-92 therapy to improve persistence 87. Allan DSJ, Chakraborty M, Waller GC, et al.
and anti-tumor activity. Theranostics 2023;13: Systematic improvements in lentiviral transduction
1506-19. of primary human natural killer cells undergoing ex
76. Xiong Q, Zhang H, Ji X, et al. A novel membrane- vivo expansion. Mol Ther Methods Clin Dev
bound interleukin-2 promotes NK-92 cell 2021;20:559-71.
persistence and anti-tumor activity. Oncoimmu- 88. Igarashi T, Wynberg J, Srinivasan R, et al. Enhanced
nology 2022;11:2127282. cytotoxicity of allogeneic NK cells with killer
77. Sahm C, Schönfeld K, Wels WS. Expression of IL- immunoglobulin-like receptor ligand incompatibility

© Chinese Journal of Cancer Research. All rights reserved. www.cjcrcn.org Chin J Cancer Res 2024;36(1):1-16
Chinese Journal of Cancer Research, Vol 36, No 1 February 2024 15

against melanoma and renal cell carcinoma cells. bioimaging of natural killer (NK) cell therapeutics.
Blood 2004;104:170-7. Biomaterials 2019;221:119418.
89. Lin W, Voskens CJ, Zhang X, et al. Fc-dependent 101. Wendel M, Galani IE, Suri-Payer E, Cerwenka A.
expression of CD137 on human NK cells: insights Natural killer cell accumulation in tumors is
into “agonistic” effects of anti-CD137 monoclonal dependent on IFN-gamma and CXCR3 ligands.
antibodies. Blood 2008;112:699-707. Cancer Res 2008;68:8437-45.
90. Denman CJ, Senyukov VV, Somanchi SS, et al. 102. Chen EB, Zhou ZJ, Xiao K, et al. The miR-561-
Membrane-bound IL-21 promotes sustained ex vivo 5p/CX 3 CL1 signaling axis regulates pulmonary
proliferation of human natural killer cells. PLoS One metastasis in hepatocellular carcinoma involving
2012;7:e30264. CX3CR1+ natural killer cells infiltration.
91. Colamartino ABL, Lemieux W, Bifsha P, et al. Theranostics 2019;9:4779-94.
Efficient and robust NK-cell transduction with 103. Galland S, Vuille J, Martin P, et al. Tumor-derived
baboon envelope pseudotyped lentivector. Front mesenchymal stem cells use distinct mechanisms to
Immunol 2019;10:2873. block the activity of natural killer cell subsets. Cell
92. Nanbakhsh A, Best B, Riese M, et al. Dextran Rep 2017;20:2891-905.
enhances the lentiviral transduction efficiency of 104. Terrén I, Orrantia A, Vitallé J, et al. NK cell
murine and human primary NK cells. J Vis Exp metabolism and tumor microenvironment. Front
2018:13;55063. Immunol 2019;10:2278.
93. Pfefferle A, Huntington ND. You have got a fast 105. Müller N, Michen S, Tietze S, et al. Engineering
CAR: Chimeric antigen receptor NK cells in cancer NK cells modified with an EGFRvIII-specific
therapy. Cancers (Basel) 2020;12:706. chimeric antigen receptor to overexpress CXCR4
94. Robbins GM, Wang M, Pomeroy EJ, et al. Nonviral improves immunotherapy of CXCL12/SDF-1α-
genome engineering of natural killer cells. Stem Cell secreting glioblastoma. J Immunother 2015;38:
Res Ther 2021;12:350. 197-210.
95. Carlsten M, Levy E, Karambelkar A, et al. Efficient 106. Ng YY, Tay JCK, Wang S. CXCR1 expression to
mRNA-based genetic engineering of human NK improve anti-cancer efficacy of intravenously
cells with high-affinity CD16 and CCR7 augments injected CAR-NK cells in mice with peritoneal
rituximab-induced ADCC against lymphoma and xenografts. Mol Ther Oncolytics 2019;16:75-85.
targets NK cell migration toward the lymph node- 107. Tong L, Jiménez-Cortegana C, Tay AHM, et al.
associated chemokine CCL19. Front Immunol NK cells and solid tumors: therapeutic potential and
2016;7:105. persisting obstacles. Mol Cancer 2022;21:206.
96. Ingegnere T, Mariotti FR, Pelosi A, et al. Human 108. Walle T, Kraske JA, Liao B, et al. Radiotherapy
CAR NK cells: A new non-viral method allowing orchestrates natural killer cell dependent antitumor
high efficient transfection and strong tumor cell immune responses through CXCL8. Sci Adv 2022;
killing. Front Immunol 2019;10:957. 8:eabh4050.
97. Muñoz-López M, García-Pérez JL. DNA 109. Fitzgerald AA, Wang S, Agarwal V, et al. DPP
transposons: nature and applications in genomics. inhibition alters the CXCR3 axis and enhances NK
Curr Genomics 2010;11:115-28. and CD8+ T cell infiltration to improve anti-PD1
98. Li Y, Hermanson DL, Moriarity BS, et al. Human efficacy in murine models of pancreatic ductal
iPSC-derived natural killer cells engineered with adenocarcinoma. J Immunother Cancer 2021;9:
chimeric antigen receptors enhance anti-tumor e002837.
activity. Cell Stem Cell 2018;23:181-92.e5. 110. Jang ES, Shin JH, Ren G, et al. The manipulation of
99. Nguyen DN, Roth TL, Li PJ, et al. Polymer- natural killer cells to target tumor sites using
stabilized Cas9 nanoparticles and modified repair magnetic nanoparticles. Biomaterials 2012;33:
templates increase genome editing efficiency. Nat 5584-92.
Biotechnol 2020;38:44-9. 111. Lu C, Guo C, Chen H, et al. A novel chimeric PD1-
100. Kim KS, Han JH, Park JH, et al. Multifunctional NKG2D-41BB receptor enhances antitumor activity
nanoparticles for genetic engineering and of NK92 cells against human lung cancer H1299

© Chinese Journal of Cancer Research. All rights reserved. www.cjcrcn.org Chin J Cancer Res 2024;36(1):1-16
16 Guo et al. CAR-NK cells: Characteristics and barriers

cells by triggering pyroptosis. Mol Immunol 2020; checkpoints for cancer immunotherapy. Chin J
122:200-6. Cancer Res 2022;34:460-82.
112. Romee R, Schneider SE, Leong JW, et al. Cytokine 121. Liang Y, Liu X, Li K, et al. Current situation of
activation induces human memory-like NK cells. programmed cell death protein 1/programmed cell
Blood 2012;120:4751-60. death ligand 1 inhibitors in advanced triple-negative
113. Lizana-Vasquez GD, Torres-Lugo M, Dixon RB, breast cancer. Chin J Cancer Res 2022;34:117-30.
et al. The application of autologous cancer 122. Ghaedrahmati F, Esmaeil N, Abbaspour M.
immunotherapies in the age of memory-NK cells. Targeting immune checkpoints: how to use natural
Front Immunol 2023;14:1167666. killer cells for fighting against solid tumors. Cancer
114. Chang Y, Jin G, Luo W, et al. Engineered human Commun (Lond) 2023;43:177-213.
pluripotent stem cell-derived natural killer cells with 123. Wang F, Wu L, Yin L, et al. Combined treatment
PD-L1 responsive immunological memory for with anti-PSMA CAR NK-92 cell and anti-PD-L1
enhanced immunotherapeutic efficacy. Bioact Mater monoclonal antibody enhances the antitumour
2023;27:168-80. efficacy against castration-resistant prostate cancer.
115. Töpfer K, Cartellieri M, Michen S, et al. DAP12- Clin Transl Med 2022;12:e901.
based activating chimeric antigen receptor for NK 124. André P, Denis C, Soulas C, et al. Anti-NKG2A
cell tumor immunotherapy. J Immunol 2015;194: mAb is a checkpoint inhibitor that promotes anti-
3201-12. tumor immunity by unleashing both T and NK cells.
116. Cytlak UM, Dyer DP, Honeychurch J, et al. Cell 2018;175:1731-43.e13.
Immunomodulation by radiotherapy in tumour 125. Elia I, Haigis MC. Metabolites and the tumour
control and normal tissue toxicity. Nat Rev Immunol microenvironment: from cellular mechanisms to
2022;22:124-38. systemic metabolism. Nat Metab 2021;3:21-32.
117. Galluzzi L, Humeau J, Buqué A, et al. Immuno- 126. Luo W, Qiu J, Zheng L, et al. Novel therapies
stimulation with chemotherapy in the era of immune targeting hypoxia mechanism to treat pancreatic
checkpoint inhibitors. Nat Rev Clin Oncol cancer. Chin J Cancer Res 2021;33:216-31.
2020;17:725-41. 127. Wang J, Lupo KB, Chambers AM, et al. Purinergic
118. Liu J, Yu Y, Liu C, et al. Combinatorial regimens of targeting enhances immunotherapy of CD73+ solid
chemotherapeutic agents: A new perspective on tumors with piggyBac-engineered chimeric antigen
raising the heat of the tumor immune microenviron- receptor natural killer cells. J Immunother Cancer
ment. Front Pharmacol 2022;13:1035954. 2018;6:136.
119. Klapdor R, Wang S, Morgan MA, et al. NK cell- 128. Yu L, Liu X, Wang X, et al. TIGIT+ TIM-3+ NK
mediated eradication of ovarian cancer cells with a cells are correlated with NK cell exhaustion and
novel chimeric antigen receptor directed against disease progression in patients with hepatitis B virus-
CD44. Biomedicines 2021;9:1399. related hepatocellular carcinoma. Oncoimmunology
120. Qian Y, Yang T, Liang H, et al. Myeloid 2021;10:1942673.

Cite this article as: Guo F, Zhang Y, Cui J. Manufacturing


CAR-NK against tumors: Who is the ideal supplier? Chin J
Cancer Res 2024;36(1):1-16. doi: 10.21147/j.issn.1000-
9604.2024.01.01

© Chinese Journal of Cancer Research. All rights reserved. www.cjcrcn.org Chin J Cancer Res 2024;36(1):1-16

You might also like