Open Access

IPSC‑derived NK cells for immunotherapy and therapeutic perspective (Review)

  • Authors:
    • Xiyao Wei
    • Chen Su
    • Yueyang Liu
    • Ningbo Wei
    • Kexin Xiang
    • Qijun Qian
    • Zenghui Xu
  • View Affiliations

  • Published online on: June 3, 2025     https://doi.org/10.3892/mmr.2025.13587
  • Article Number: 222
  • Copyright: © Wei et al. This is an open access article distributed under the terms of Creative Commons Attribution License.

Metrics: Total Views: 0 (Spandidos Publications: | PMC Statistics: )
Total PDF Downloads: 0 (Spandidos Publications: | PMC Statistics: )


Abstract

Natural killer (NK) cell‑based immunotherapy has emerged as a transformative approach for cancer treatment However, its widespread clinical application faces several challenges, such as donor variability, limited scalability and functional heterogeneity of primary NK cells. Additionally, issues including in vivo persistence, resistance to tumor microenvironment and safety concerns related to genomic instability further hinder its clinical application. Induced pluripotent stem cell (iPSC)‑derived NK cells offer a promising solution. They provide high homogeneity and quality control, genetic engineering flexibility and inexhaustible cell source. This present review highlighted the unique advantages of iPSC‑ NK cells, including clonal uniformity, enhanced cytotoxicity and suitability for large‑scale production, positioning them as an ideal ‘off‑the‑shelf’ therapeutic platform. It discussed the biological properties of iPSC‑derived NK cells, advances in differentiation protocols and strategies to augment their anti‑tumor efficacy through genetic engineering, such as chimeric antigen receptor integration and cytokine optimization. Despite these advantages, several challenges remain, including the need to optimize differentiation efficiency, ensure the safety of gene editing (such as off‑target effects) and improve the in vivo migration and infiltration abilities. With technological advances and clinical validation, this present review aimed to guide future research toward overcoming these barriers to clinical implementation. Ultimately, it is expected that iPSC‑NK will become a core means of next‑generation immunotherapy, promoting the combination of personalized and inclusive cancer treatment.

Introduction

As the first line of defense against infections and malignancies, human natural killer (NK) cells are a critical component of innate immune system. Unlike T cells, which require priming, NK cells can eliminate virus-infected cells and transformed cells through immune mechanisms (1). Various NK cell sources have been investigated in clinical trials for cellular immunotherapy (2). Induced pluripotent stem cel -derived NK cells (IPSC-NK) have emerged as a research hotspot in tumor immunotherapy due to their scalability, standardized manufacturing potential and genetic engineering capabilities. Compared with other immunotherapies, NK cells demonstrate improved cytotoxic activity and are less sensitive to tumor immune escape strategies, making them a promising approach for cancer treatment (3).

NK cells do not require human leukocyte antigen (HLA) compatibility, reducing the risk of complications such as graft-versus-host disease (GVHD) and cytokine release syndrome (CRS), even when administered as allogeneic cells (4). Their favorable safety profile, coupled with notable anti-tumor capabilities, positions NK cells as a compelling cellular candidate for the application of chimeric antigen receptor (CAR) technology (5). This enables the strategic redirection of their cytotoxic capabilities towards precise targets (6,7).

Despite these advantages compared with other immunotherapies, NK cell-based therapies face several challenges, including immunosuppressive tumor microenvironments, limitations in cell manufacturing and insufficient therapeutic persistence. IPSC-NK cells have emerged as an attractive alternative to overcome these limitations (8). IPSC-NK cells facilitate the mass production of homogeneous NK cells, which can be stored for later use. Moreover, both viral and non-viral methods can be used to effectively genetically modify these cells to meet the needs of different cancer treatments (9). Therefore, in biomedical research and clinical applications, iPSCs hold great promise for translational research and clinical applications (10). As the ‘off the shelf’ product, iPSC-NK cells possess strong drug-forming characteristics and represent a promising strategy in cancer immunotherapy. This present review examined the immunotherapy and therapeutic perspective of NK cells derived from iPSCs.

Biological properties of NK cells

Unlike T and B cells, NK cells lack genetically rearranged antigen receptors, allowing them to directly eliminate target cells without prior sensitization (11). NK cells are positioned as promising candidates for cancer adoptive cell therapy because of this unique feature, as well as their ‘off-the-shelf’ availability and low risk of GVHD (12,13). NK cells are a type of lymphocyte that play a crucial role in the innate immune system. Their development begins in the bone marrow, where hematopoietic stem cells differentiate into common lymphoid progenitors. These progenitors further differentiate into NK cell precursors by downregulating CD34 and upregulating CD56, with IL2RB (CD122) expression marking the entry into the NK cell lineage (14). The precursors migrate to lymph nodes, where cytokines from stromal and dendritic cells facilitate their maturation into functional NK cells. This process is characterized by the differential expression of genes, including CD34, KIT, KLRB1, CD244 and interleukin (IL)-15R (1517). During this process, NK cells start expressing receptors that enable them to recognize and bind to target cells, such as infected or cancerous cells.

NK cells are broadly categorized into CD56bright and CD56dim NK subsets (Fig. 1). The CD56dim subset constitutes ~90% of peripheral blood NK cells and plays a primary role in direct target cells elimination through the release of perforin and granzymes. These cells exhibit the strongest cytotoxic activity and antibody-dependent cell-mediated cytotoxicity (ADCC) capabilities (1820).

By contrast, CD56bright cells, which represent 2–10% of peripheral blood NK cells, have less cytotoxic activity but play a crucial role in immunomodulation. They can interact with dendritic cells (DCs) and T cells to maintain the balance and effectiveness of the immune system (21). Activated NK cells secrete cytokines such as interferon (IFN)-γ, tumor necrosis factor (TNF)-α and granulocyte-macrophage colony-stimulating factor to enhance cytotoxic T cell responses and macrophage antigen presentation. NK cells can be activated by cytokines secreted by DCs, such as IL-12 and IL-18 (22,23).

CD56bright cells also exert anti-proliferative, anti-angiogenic and pro-apoptotic effects on cancer cells while recruiting DCs to the tumor microenvironment via chemokines such as C-C motif chemokine ligand 5, recombinant chemokine C-Motif ligand (XCL) 1 and XCL2, thereby promoting anti-tumor immunity (24). Furthermore, CD56bright cells express chemokine receptors such as C-C chemokine receptor type (CCR) 7, CCR5 and CXCR4, which allow them to migrate and localize to secondary lymphoid organs. They also express CD62L-selectin, which interacts with high endothelial venues to facilitate homing and retention in lymphoid tissues (17,25,26). This characteristic underscores the crucial role of NK cells in immune regulation and cytokine-mediated immune responses (25).

Adoptive NK cell therapy

The unique biological properties of NK cells provide a strong rationale for their application in adoptive immunotherapy. NK cell activation and inhibition are governed by a range of activating and inhibitory receptors (Table I) (13). Whether NK cells are activated or inhibited depends on how signals from these receptors interact (27). The capacity of NK cells to detect and eliminate cancer cells through mechanisms such as MHC-I downregulation recognition is a defining characteristic of their activity (28,29). This recognition is facilitated by the presence of killer cell immunoglobulin-like receptors (KIRs) on NK cells, which are sensitive to the absence of major histocompatibility complex (MHC) class I molecules (30). Inhibitory KIRs contain immune receptor tyrosine-based inhibitory motifs, which mediate inhibitory signals, while activating KIRs associate with immunoreceptor tyrosine-based activation motifs, transmitting activating signals (31).

Table I.

Human NK cell receptors and function.

Table I.

Human NK cell receptors and function.

ReceptorFunctionLigands on tumor
NKG2DActivationMIC-A/B, Rael, H60, ULBP
CD94/,NKG2CActivationHLA-E
NKP30ActivationHLA-EB7-H6, BAT3, CMVPP65
NKP44ActivationViral HA
NKP46ActivationViral HA
CD160ActivationHLA-C
KIR2DS1ActivationHLA-C
CD94/NKG2AInhibitionHLA-E
CD96InhibitionCD155
LAIR1InhibitionCollagen
KLRG1InhibitionE-Cad
PD-1InhibitionPD-L1/2
SIGLEC-3/7/9InhibitionSialic acid
NKR-PIAInhibitionCLEC2D
TIGITInhibitionCD155
iKIRsInhibitionHLA-I
ILT2InhibitionHLA-C
CD96InhibitionCD155

[i] NK, natural killer; MIC-A/B, MHC class I polypeptide-related sequence A/B; HLA-E, major histocompatibility complex; BAT3, HLA-B associated transcript 3; CMVPP65, cytomegalovirus phosphoprotein 65; HA, hemagglutinin; CLEC2D, C-Type lectin domain family 2 member D; PD-L1/2, programmed death-1; KIR2DS1, killer cell immunoglobulin-like receptor, two Ig domains and short cytoplasmic tail 1; LAIR1, leukocyte-associated immunoglobulin-like receptor 1; KLRG1, killer cell lectin-like receptor subfamily G member 1; SIGLEC-3/7/9, sialic acid-binding immunoglobulin-like lectin 3/7/9; NKR-PIA, killer cell lectin-like receptor subfamily B member 1; TIGIT, T cell immunoglobulin and ITIM domain; iKIR, inhibitory killer-cell immunoglobulin-like receptors; ILT2, immunoglobulin-like transcript 2.

When KIRs on NK cells encounter and bind to MHC class I molecules on the surface of healthy cells, inhibitory signals are transmitted, preventing the NK cells from attacking these healthy cells (32,33). However, in the case of aberrant expression of MHC class I molecules, the inhibitory receptors on NK cells are unable to recognize the altered MHC class I molecules, resulting in an inability to transmit inhibitory signals (Fig. 2) (34). As a result, when the activating signal outweigh inhibitory signals, NK cells receive an activating signal and proceed to recognize and eliminate the tumor cells. For example, when tumor cells downregulate MHC class I molecule expression, inhibitory signals on NK cells are lifted, releasing the ‘missing-self’ restraint and activating their cytotoxic functions. Simultaneously, MHC-I-deficient tumor cells may upregulate stress-induced molecules, which bind to activating receptors on NK cells, further amplifying cytotoxic signaling (35). This mechanism enables NK cells to target tumors that evade T cell-mediated immune surveillance, forming a complementary anti-tumor immune defense (36). human leukocyte antigen (HLA)-E-related peptides transmit inhibitory signals by interacting with NK inhibitory receptors (37).

For instance, NKG2A dimerizes with CD94 on the cell surface and binds to HLA-E, which is crucial for tumors to resist immune cell activity. In hepatocellular carcinoma (HCC) tissues, the surface expression of NKG2A is increased in NK cells, together with the expression of its corresponding ligand, HLA-E (38,39). Analysis of gene expression data in tumor samples reveals a significant correlation between HLA-E expression levels within tumor tissues and expression levels of NKG2A and CD94 on the surface of NK cells (40). Unlike T cell activation, which requires dual signaling, the activation of NK cells is determined solely by the regulation of surface receptors and is not constrained by MHC. As the tumor progresses, NK cells are rapidly activated, executing their functions ahead of T cells (41,42).

Additionally, NK cells can eliminate tumor cells through ADCC, mediated by Fc γ receptors, which engage antibodies to induce target cell death (4345). The most important activation receptor in this process is CD16, an antibody receptor that binds immunoglobulin Fc, working alongside other key activation receptors such as NKG2D and natural cytotoxicity receptor (NCR) (46,47). A regulatory mechanism for NK cell cytotoxicity is also provided by the binding of ligands on target cell surfaces to NCR family receptors, including NKp30, NKp44, NKp46 and NKG2D (48,49).

For subsequent gene editing of iPSC-NK cells, it is crucial to understand the characteristics and functions of activating and inhibitory receptors on the surface of NK cells. By exploiting gene editing technology, the expression of receptors on the surface of NK cells can be precisely regulated, thereby optimizing the balance between their activating and inhibitory signals. This approach can enhance the anti-tumor and anti-viral capabilities of NK cells while reducing the risk of potential immune overreaction.

Sourcing NK cells

Due to the limited antitumor effect of autologous NK cells, researchers have shifted their focus from autologous to allogeneic NK cell therapy (50). Over the past decade, various NK cell sources have been investigated in clinical trials for cellular immunotherapy, including peripheral blood, umbilical cord blood and NK-92 (Fig. 3). NK cells sourced from peripheral blood are easy to acquire and handle. Each of these sources has distinct advantages. Umbilical cord blood provides a readily available source with weaker allogenic reactions and a low risk of viral transmission (51). Meanwhile, NK-92 cells, an infinitely proliferating cell line, exhibit high cytotoxicity and stable immune properties (52). However, each of these sources has drawbacks. Peripheral blood and umbilical cord blood-derived NK cells are inconvenient to store and subject to donor variability. NK-92 cells, while highly cytotoxic, pose potential carcinogenic risks, lack CD16 and NKp44 and require irradiation for inactivation, which markedly reduces their proliferative capacity and cytotoxicity. The absence of CD16 also hampers their ability to perform ADCC (53,54).

To address these limitations, IPSC-NK cells have emerged as a promising alternative, which can be used as a standardized, ‘off-the-shelf’ allogeneic cell therapy for treating diverse malignancies and amenable to genome editing (55). Furthermore, modified iPSC-NK cells have shown high target specificity, persistence and immune activation potential in cancer treatment (8,56).

Development and differentiation of iPSC-derived NK cells

The development of iPSC technology began in 2006. The approach was pioneered by Shinya Yamanaka, who developed a method to generate iPSCs by introducing four transcription factors (OCT3/4, SOX2, KLF4 and c-Myc) into somatic cells (5759). IPSCs show characteristics identical to embryonic stem cells (ESCs) in terms of morphology, proliferation, gene expression and the ability to form teratomas.

IPSC-NK cell therapy is a novel cancer immunotherapy approach based on iPSC technology. In 2024, Kiran et al (60) developed a method to induce and sustain transgene-free human iPSCs, enabling efficient and uniform amplification using reprogramming factors such as SOX2, OCT4, KLF4, MYC, NANOG, LIN28 and SV40 T antigen in a feeder-free system. This method showed an effective performance against malignant brain rhomboid tumor cells. To effectively mitigate the risk of residual exogenous genes, technologies focus on employing non-integrating vectors and small molecule compounds to achieve gene silencing or post-reprogramming removal in iPSCs (61). The differentiation of iPSC into mature NK is typically divided into three stages. Initially, to promote the differentiation of iPSCs into hematopoietic progenitor cells (HPCs), they are co-cultured with a combination of small molecules and cytokines, or irradiated stromal cell lines. Subsequently, CD34+ HPCs are isolated and enriched before being directed towards NK cell differentiation using specific cytokines (IL-3, IL-7, IL-15, SCF and FLT3L) or through co-culture with a second stromal cell line. In the final stage, iPSC-NK cells co-cultured with irradiated and engineered feeder cells to further expand their population (62,63).

In addition, researchers formulated a technique for spin embryoid body (EB) protocol, which has further enhanced the efficiency of iPSC differentiation (64,65). The differentiation of iPSCs into NK cells occurs through a multi-stage process: First, iPSCs are co-cultured with small molecules and induction media, which promotes their differentiation into HPCs. This stage typically takes 12 days to produce CD34+ HPCs. Second, lymphoid induction media and differentiation factors are used to induce lymphoid progenitor cells. The third stage involves guiding the differentiation of iPSCs into NK cells using specific NK induction media. The fourth and fifth stages involve the maturation process of NK cells (Fig. 3).

Zhang et al (66) confirmed the successful differentiation process from iPSC to iPSC-NK cells using EBs and analyzed the temporal changes in the expression of key genes through bulk RNA-seq analysis. Their findings revealed that while iPSC-NK cells share transcriptomic similarities with PBMC-derived NK cells, they maintained unique phenotypes characteristics. Pluripotency genes are highly expressed at the iPSC stage but gradually decreased with differentiation, becoming barely detectable at the EB stage. Hematopoietic-related genes were expressed at the EB stage and gradually increased during the differentiation process. Gene's specific to NK cells, such as GZMB, PRF1 and IL2RB, were gradually expressed from the early stage of differentiation and peaked in the late stage. The expression of some NK cell-specific markers (such as NKG2D, NKp46 and NKp30) began at the early stage of in the process of EB differentiation into iPSC-NK cells. During the middle stage, there was an increase in the expression of GATA2, the HSC regulator and key regulator of NK cell maturation. Simultaneously, the expression of mature NK cell-specific markers (such as natural killer cell cytotoxicity receptors and activation receptors) gradually increased. Compared with traditional 2D culture, EB-based iPSC-NK cells improved imitate the in vivo microenvironment. combining Clinical-grade iPSC-NK manufacturing is made possible by the cost reduction that results from combining EB with bioreactor technology (66).

Utilization of iPSC-derived NK cell in cancer treatment

In recent years, iPSC-NK cells have emerged as a novel direction in the field of immunotherapy. These cells open new possibilities for treating various diseases, particularly cancer, by harnessing the advantages of iPSCs and NK cells. They have demonstrated significant clinical benefits in cancer treatment, as well as in autologous and allogeneic transplantation, infectious diseases, antiviral therapy and autoimmune diseases (66,67).

Currently, antibodies are extensively utilized in numerous cancer treatment modalities. However, treatment with antibodies alone might not be enough to increase the immune response because certain tumor patients (both treated and untreated) have severe lymphopenia (68). As a result, the infusion of a substantial quantity of NK cells is often required in the most clinical trials involving NK cells, ranging from 5×106−1×108 cells/kg (8). One major advantage of iPSC-NK cells is their ability to undergo genetic modification and cryopreservation after differentiation, facilitating the production of homogeneous functional NK cells at a clinical scale (69). A study showed that differentiated iPSC-NK cells exhibit remarkable 3,000-fold expansion when Compared with primary NK cells Over 200 doses, each containing >1×109 cells, can be produced by utilizing this cell production scale (70). Markedly, iPSC-NK cells retain the characteristic NK cell phenotype, including the presence of activating receptors such as NKG2D, NKp44, NKp46 and DNAM-1 (66).

Furthermore, a study analyzing NK cell populations from peripheral blood, umbilical cord blood and iPSC-NK cells found that these different sources exhibit relatively similar expression patterns of cell surface antigens, as well as activation and inhibitory receptors. Notably, unlike CB-NK and PB-NK cells, iPSC-NK cells show variable expression of killer immunoglobulin-like receptors (KIRs). However, their cytotoxic activity against tumor targets is not markedly impaired by this mechanism. In fact, Compared with PB-NK cells, iPSC-NK cells produced more IFN-γ and TNF-α, thereby enhancing their function (65).

In November 2018, the FDA approved FT500, the first clinical trial for iPSC-NK cell immunotherapy (56). Recent clinical trials have shown that iPSC-NK cells boost anti-tumor cytotoxicity and promote T cell activation and homing (71). Higher cytotoxicity against lung carcinoma cells, hepatocyte carcinoma cells, ovarian adenocarcinoma cells, melanoma cells and myeloid leukemia cells is demonstrated by their ability to recognize and lyse HLA-I downregulated tumor cells. This is particularly beneficial for solid tumor patients receiving anti-programmed death-1 (PD-1) or anti- programmed death ligand 1 (PD-L1) antibody therapy (72). Moreover, iPSC-NK cells may be cryopreserved and retained for subsequent repeated infusions.

A study comparing fresh, frozen and thawed PB-NK cells showed that among patients who received cell therapy, frozen cells did not proliferate and exhibited reduced cytotoxicity after thawing. By contrast, iPSC-NK cells can be cryopreserved at high densities. For example, both the FT596 and FT396 clinical trials used a density of 1.1×108 for cryopreservation and the thawed cells still demonstrated high recovery rates and cytotoxicity (73). The method of producing cGMP-grade NK cells from iPSC lines is a significant advance. This approach not only improves NK cell production efficiency but also preserves their functional integrity. Additionally, iPSC-NK cells serve as a powerful tool for cancer treatment, serving as a cell seed bank (62,74). While NK cells exhibit advantages compared with other immunotherapies, several limitations for solid tumors should also be overcome. The following sections introduce the strategies to enhance the function of iPSC-NK cells (Fig. 4).

Effects of cytokines and chemokines on iPSC-derived NK cells

IL-15 plays a pivotal role in the immune system, particularly in the development, maintenance and function of NK cells (75,76). IL-15 induces the proliferation of NK cells through binding to its receptor and activation of downstream signaling pathways, such as JAK-STAT and PI3K/AKT (77,78). Research has shown that culturing NK cells with a combination of IL-15, IL-18 and IL-12 enhances their targeting and killing of tumor cells both in vivo and in vitro (79). For iPSC-NK cells, Chen et al (80) developed a TALEN-based workflow to knock in IL-15, which enhanced the cellular function and persistence of iPSC-NK cells (80). A study by Kim et al (45) demonstrate that NK-exosIL-15/21 (natural killer cell-derived exosomes loaded with IL-15 and IL-21) enhances cytotoxicity and apoptotic activity in Hep3B cells. This effect was achieved by activating specific pro-apoptotic proteins, including Bax, cleaved caspase-3, cleaved poly ADP-ribose polymerase, perforin and granzyme B. Additionally, the treatment inhibited the anti-apoptotic protein Bcl-2, which prevents apoptosis and promotes cell survival.

Furthermore, a platform has been developed using CISH knock out in iPSC-NK cells to enhance JAK-STAT signaling through IL-15. Through improved metabolic fitness, which is characterized by mTOR signaling, this alteration directly contributes to enhanced NK function (11). These mechanisms still need to be deeply explored to optimize treatment strategies and translate pre-clinical findings into more effective clinical therapies for cancer patients (81).

Increased CD16 receptors on iPSC-derived NK cells

CD16 is a receptor involved in ADCC (82). When CD16a recognizes IgG-coated targets, NK cells release various cytotoxic molecules to mediate the death of the target cells (83,84). The cleavage of CD16a by ADAM17 (a disintegrin and metalloproteinase) is one of the mechanisms underlying its shedding. Activated NK cells exhibit a loss of CD16, which is cleaved by the ADAM17, and the homing receptor CD62L (84,85). Therefore, a strategy to improve cellular ADCC is to target ADAM17 to prevent CD16a shedding (85,86).

Research has found that using CRISPR/Cas9 technology to knock out ADAM17 can lead to improved production of IFN-γ and enhanced NK cell activity both in vitro and in vivo (87). Clinical trials have modified iPSC-NK cells to express a novel high-affinity 158V, non-cleavable CD16 Fc downregulation and enhance their binding ability with monoclonal antibodies (19). Meng et al (86) developed a novel hnCD16 fusion receptor, which consist of the extracellular domain of hnCD16, NK cell-specific co-stimulatory molecules (2B4 and DAP10) and the intracellular domain of CD3ζ. In vitro, iPSC-NK cells with hnCD16 fusion receptor demonstrate a marked increase in cytokine secretion against tumor cells compared with the control group. Additionally, it has been demonstrated that the anti-tumor capability of iPSC-NK cells can be markedly enhanced by combing high-affinity, non-cleavable CD16 fusion receptor (hnCD16FR)-iPSC-NK cells with CD20 monoclonal antibodies (88).

Increased other receptors on iPSC-derived NK cells to enhance functions

CD38 is a transmembrane protein primarily responsible for catalyzing the activity of molecules such as nicotinamide adenine dinucleotide and nicotinamide mononucleotide (89). Of CD38, ~90% is located on the cell membrane. To enhance the cytotoxic efficacy of NK cells, a strategic approach involves suppressing CD38 activity through genetic ablation (54,90).

Research has shown that knocking out CD38 and introducing the CD16-158V receptor in NK cells results in stronger ADCC effects and improved anti-tumor activity, particularly in multiple myeloma (90). NKG2A is an inhibitory receptor that binds to HLA-E on NK cells. Qin et al (68) focus on delating the expression of NKG2A by knocking out the NKG2A receptor gene in iPSC-NK cells, demonstrating excellent killing ability against tumors that highly express HLA-E.

T cell immune receptor with immunoglobulin and ITIM domains (TIGIT) and CD73 are key molecules in immune inhibitory pathways, often highly expressed in the tumor microenvironment, thereby suppressing immune cell function (68). Targeting TIGIT and CD73 alleviates immune suppression, enhancing anti-tumor immune responses. In glioblastoma therapy, inhibiting the TIGIT and CD73 pathways helps restore the activity of NK cells and T cells, leading to more effective tumor cell killing (91,92). Lupo et al (93) used the synNotch system to program iPSC-NK cells to express molecules capable of disrupting TIGIT and CD73 activity, showing a good performance in disrupting the immunosuppressive network in glioblastoma (93). Advances in gene editing technology have enabled the reduction of immunosuppression by inactivating HLA genes. This is achieved by the deletion of their common component, β-2 microglobulin (B2M), using CRISPR-Cas9 (94).

By employing gene editing techniques, it is possible to generate cell lines with a consistent genetic background. This approach minimizes phenotypic variations arising from genetic disparities, thereby enhancing the precision and reliability of disease research.

Increased targeting on iPSC-derived NK cells to enhance functions

Currently, researchers are improving the targeting capabilities of iPSC-NK cells by modifying them with CAR. A classic CAR consists of an extracellular recognition domain, such as a single-chain variable fragment (scFv) that recognizes tumor-specific antigens, along with a transmembrane domain and an intracellular signaling domain (10,95). The remarkable clinical efficacy of CAR-NK cells has sparked significant enthusiasm among researchers worldwide. As a result, clinical trials utilizing CAR-NK cells derived from diverse sources are currently underway at a rapid pace (Table II) (96). However, genetic modification of primary NK cells presents several challenges. The freeze-thaw process further complicates these challenges by compromising both the viability and anti-tumor capabilities of these cells (78). Additionally, the expansion and persistence of CAR-NK cells are limited by metabolic exhaustion and insufficient cytokine support within the tumor microenvironment (79).

Table II.

CAR-NK cell clinical trials.

Table II.

CAR-NK cell clinical trials.

Number of NCTStart yearStageTumorsTargetNK sourceLocations
NCT063070542024/2/28Phase 1Relapsed Adult AML. Refractory AMLCLL-1PB-NKShanghai General Hospital, Shanghai Jiao Tong University School of Medicine
NCT062422492024/1/27Phase1/Phase 2Multiple Myeloma, RefractoryBCMAPB-NKShahid Beheshti University of Medical Sciences
NCT062012472023/12/30Early Phase 1Acute Myeloid Leukemia, in Relapse; Acute Myeloid Leukemia RefractoryCD123PB-NKPeking University People's Hospital
NCT061827352023/12/6Phase 1Renal Cell carcinomaCD70PB-NKFudan University
NCT060664242023/9/27Phase 1Solid TumorsTROP2PB-NKM.D. Anderson Cancer Center
NCT060450912023/9/13Early Phase 1Multiple Myeloma; Plasma Cell LeukemiaBCMAPB-NKShanghai Changzheng Hospital
NCT060278532023/8/30Phase 1Acute Non-Lymphoblastic Leukemia; Myeloid Leukemia; Acute Myeloid Leukemia; Acute Graft Versus Host DiseaseCLL1iPSC-NKZhejiang University
NCT060064032023/8/17Phase 1/Phase 2AML; Blastic Plasmacytoid Dendritic Cell Neoplasm; relapse Leukemia; Refractory LeukemiaCD123PB-NKChongqing Precision Biotech Co., Ltd
NCT059876962023/7/24Phase 1AML, Adult; Minimal Residual DiseaseCLL1/CD33iPSC-NKInstitute of Hematology & Blood Diseases Hospital, China
NCT059229302023/6/20Phase 1/Phase 2Pancreatic Cancer; Ovarian cancer; AdenocarcinomaTROP2CB-NKM.D. Anderson Cancer Center
NCT058566432023/5/3Early Phase 1Ovarian Epithelial CarcinomaSZ-011PB-NKShantou University Medical College
NCT058455022023/4/25Not ApplicableAdvanced Hepatocellular CarcinomaSZ003 Shantou University Medical College
NCT058427072023/3/25Phase 1/Phase 2Refractory or Relapsed B-cell Non-Hodgkin lymphomaCD19/CD70CB-NKShanghai Tongji Hospital, Tongji University School of Medicine
NCT057763552023/3/8Not ApplicableOvarian CancerNKG2DPB-NKHangzhou Cheetah Cell Therapeutics Co., Ltd
NCT057392272023/2/11Early Phase 1Acute lymphoblastic Leukemia; B-cell Lymphoma; Chronic Lymphocytic LeukemiaCD19PB-NKXuzhou Medical University
NCT057475862023/1/30Not ApplicableMultiple Myeloma in Relapse; Multiple Myeloma, RefractoryBCMAPB-NKZhejiang University
NCT057348982023/1/30Not ApplicableAcute Non-Lymphoblastic leukemia; Acute Myeloid Leukemia;NKG2DPB-NKZhejiang University
ChiCTR230007662023/9/21Early Phase 1AMLNKG2DPB-NKTongji Hospital, Tongji Medical College, Huazhong University of Science and Technology

[i] CAR, chimeric antigen receptor; NK, natural killer; PB, peripheral blood; CB, cord blood.

By contrast, iPSC-NK cells offer several advantages, including a low requirement for seed cells, scalability, cost-effectiveness and the potential for autologous supply with minimal immunogenicity. Furthermore, these cells can be cryopreserved long-term, ensuring immediate availability for critically patients (9,76,77). Consequently, the development of CAR-iPSC-NK cells has emerged as a promising strategy for generating readily available allogeneic lymphocytes that can specifically target and combat malignant tumors. At present, CAR-iPSC-NK cells have evolved into a promising strategy for producing allogeneic lymphocytes to specifically target and combat certain malignant tumors (97,98). The potential of CAR-iPSC-NK cells was first reported in June 2018, when researchers introduced a CAR construct (scFv-NKG2D-2B4-CD3ζ) into iPSC-NK cells. The results demonstrated that CAR-iPSC-NK cells exhibited antigen-specific cell killing and increased expression of the granule marker CD107a (9).

In vivo experiments further show that mice treated with CAR-iPSC-NK cells experienced fewer side effects and much higher survival rates than treated with CAR-T cells. This study provided the first evidence that CAR-targeted NK cell therapy a viable option for treating refractory malignancies when chimeric antigen receptors are combined with the clinical-scale NK cells from iPSCs (9). Karvouni et al (99) developed a specific CAR targeting GPRC5D, which is highly expressed in Multiple myeloma (MM). They conducted a series of gene edits on iPSC-NK cells, including the incorporation of IL15/IL15RF fusion protein, hnCD16 and the knockout of CD38. These findings indicated that CAR-iPSC-NK cells a demonstrated strong cytotoxicity and tumor-clearing capabilities in allogeneic MM transplants (99). In vivo results indicated that two out of five mice achieved complete tumor clearance by day 80, highlighting the synergistic effect of CAR and hnCD16 (100102).

Wang et al (103) developed 70-CAR-iPSC-NK cells, incorporating four gene edits which exhibited robust cytotoxicity against a wide range of tumors. The efficacy in precisely targeting lymphoma and renal cancer was further substantiated through xenograft models. The study also found that allogeneic reactive T cells expressed high levels of CD70, which 70-CAR-iPSC-NK cells effectively targeted and cleared, enhancing their survival and persistence (103). This finding underscores the importance of selecting an appropriate CAR construct to maximize the anti-tumor efficacy of CAR-iPSC-NK cells. The importance of CAR selection is further emphasized in ovarian cancer xenograft model. Compared with PB-NK cells, iPSC -derived NK cells and T-CAR-expressing iPSC-NK cells (scFv-CD28-CD28-CD137-CD3ζ,), NK-CAR expressing-iPSC-NK (scFv-NKG2D-2B4-CD3ζ) cells demonstrated markedly improved tumor suppression and prolonged survival (9).

Additionally, studies have reported remarkable anti-tumor performance of iPSC-derived NK cells expressing various CAR constructs, such as EGFR-CAR, CD19-CAR and CD33-CAR (104106). By carefully selecting the CAR construct and employing metabolic engineering and gene-editing strategies, the functionality and targeting capabilities of CAR-iPSC-NK cells can be optimized.

Advances in clinical studies of iPSC-derived NK cells

IPSC-NK cells are currently undergoing clinical trials to evaluate their potential in treating various diseases. In order to provide data support for further clinical applications, these trials aim to determine the safety, efficacy and feasibility of iPSC-NK cells (Table III). The fastest-progressing clinical projects for iPSC-NK thus far primarily include those associated with Fate Therapeutics (FT596, FT576) and Century Therapeutics (CNTY-101).

Table III.

Ongoing clinical trials with iPSC-NK cells.

Table III.

Ongoing clinical trials with iPSC-NK cells.

CompanyiPSC platformProductIndicationsPhase
Fate TherapeuticsiNKFT576Multiple MyelomaPhase I
iNKFT522B-Cell LymphomaPhase I
CenturyiNKCYTN-101B-Cell LymphomaPhase I
iNKCYTN-104Acute Myeloid LeukemiaPreclinical
iNKCYTN-106Multiple MyelomaPreclinical
NuwacelliNKNCR300Myelodysplastic syndromesPhase I
iNKNCR301Myelodysplastic syndromePreclinical
iNKNCR305Solid tumorPreclinical
Cytovia TherapeuticsiNKCYT-103Hepatocellular carcinomaPreclinical
iNKCYT-303Hepatocellular CarcinomaPreclinical
CYT-150
CAR-iNKCYT-503Hepatocellular CarcinomaPreclinical
CAR-iNKCYT-538Multiple MyelomaPreclinical
CAR-iNKCYT-501Glioblastoma MultiformePreclinical
HebecelliNKHC101Acute Myeloid LeukemiaPreclinical

[i] iPSC-NK, induced pluripotent stem cell-derived NK cells; CAR, chimeric antigen receptor; NK, natural killer.

In February 2025, the first clinical trial of an iPSC-derived CAR-NK cells product was completed. FT596 (trial no. NCT04245722) received Investigational New Drug approval from the US FDA (107). This product includes a CD19 CAR, a high-affinity, non-cleavable CD16 Fc receptor and an interleukin-15-interleukin-15 receptor fusion (58,108,109). In the Phase I trial, FT596 demonstrated a complete Remission (CR) rate of 85% (17/20 patients) in relapsed follicular lymphoma, with a median duration of response of ~16.9 months. Among patients previously treated with CD19 CAR-T therapy, the CR rate reached 30%. Furthermore, the study found that the proportion of PD1+CD8+ T cells in the tumor was positively associated with efficacy. This suggests that FT596 may exert an auxiliary anti-tumor effect by activating endogenous T cells, achieving a dual combined action of NK cells and T cells (107).

FT522 (NCT05950334) is a further optimized version of FT596. In order to expand the range of indications and reduce dependence on preconditioning chemotherapy, it incorporates an allo-defense receptor targeting 4-1BB function. Peripheral B lymphocytes were rapidly and profoundly reduced in patients receiving FT522 therapy in Phase 1 clinical trials. FT522 demonstrated enhanced persistence in comparison to the CAR-iPSC-NK cell product of the preceding generation, FT596 (110).

FT576 (NCT05182073) is an ‘off-the-shelf’ NK cell therapy derived from iPSC lines. It has been engineered with a CAR targeting B-cell maturation antigen (BCMA) and an IL-15 receptor fusion protein. In an evaluation involving nine patients, the following observations were made: No dose-limiting toxicities, no grade CRS, no immune-related neurotoxicity, no cases of GvHD. Notably, one patient who had previously undergone five lines of treatment achieved a very good partial response after the second administration of FT576 monotherapy, accompanied by a significant decrease in soluble BCMA (102). These findings indicated iPSC-NK cells have promising safety and efficacy for treating solid tumor, offering therapeutic benefits even in patients with extensive prior treatments (100102).

CNTY-101 represents a pioneering cell therapy product candidate with six precise gene edits (111). These edits include: The incorporation of a CD19-CAR for targeted cell recognition, insertion of transgenes encoding HLA-E protein to disrupt B2M, insertion of transgenes encoding the extracellular and transmembrane domains of EGFR, implementation of Allo-Evasion™ technology (Century Therapeutics) to enhance compatibility across diverse patient populations, abnormal cells can be rapidly eliminated through the integration of suicide genes or drug-inducible switches to rapidly eliminate abnormal cells when toxicity induced by cellular therapies (94). These modifications enhance the cytotoxic effects of CNTY-101, even after over 15 successive rounds of in vitro killing. In vivo studies have shown significant effects, with fresh cells demonstrated a notable 91% reduction in tumor growth and cryopreserved cells showing a substantial 76% reduction. These results underscore the exceptional potential of CNTY-101, which is attributed to its enhanced properties and efficacy in precisely targeting and suppressing tumor growth in preclinical settings (112114).

Therefore, CAR-iPSC-NK cells can directly enhance anti-tumor activity by improving ADCC and cytokine secretion. The clinical trials of iPSC-NK cells aim to validate their potential in treating cancer and autoimmune diseases, providing scientific evidence for their widespread application as an innovative cell therapy (3,115).

Challenges of iPSC-derived NK cells for clinical application

The performance of iPSC-NK cells in treating malignancies has generated great interest in their application. However, several obstacles must be tackled before their successful use. First, quality challenges cannot be overlooked. The preparation and differentiation of iPSCs require strict control of various conditions to ensure the final cell products are consistent and of high quality (71).

Heterogeneity in differentiation protocols may yield iPSC-NK cells with divergent phenotypes and functional profiles (116). For example, compared with feeder-free differentiation strategies, a lymphoid-based differentiation strategy using OP9 cells may produce more mature and potent iPSC-NK cells (117). However, due to technical limitations and the complexity of cell biological characteristics, it is difficult to completely avoid issues such as cell variation and contamination in practical operation. Researchers should evaluate the long-term safety of cell therapy and closely monitor patients for adverse reactions and side effects when applying these cells to cancer patients (118). This will include monitoring immune responses, tumor formation and other potential risks associated with cell therapy. To enhance the efficacy and safety of iPSC-NK cells in the tumor microenvironment, future research needs to further explore how to optimize their preparation and modification methods.

Second, the tumor microenvironment plays a crucial role in enabling tumor cells to evade NK cell immune surveillance (10,119). The migration and infiltration abilities of iPSC-NK cells are crucial for targeting tumors. However, the dense stroma, high interstitial fluid pressure and abnormal vascular structures in the tumor microenvironment can impede these processes (116,120). Lactic acid in the tumor microenvironment suppresses NK cell cytotoxicity by inhibiting mTOR signaling (121123).

To counteract this effect, strategies such as engineering iPSC-NK cells with lactate dehydrogenase overexpression have been employed (124,125). Enhancing the migration and infiltration abilities of iPSC-NK cells to enable them to reach the tumor site more effectively is an important research direction. Moreover, immunosuppressive factors in the tumor microenvironment also negatively impact the activity of iPSC-NK cells (126,127). The proliferation, activation and effector functions of iPSC-NK cells can be inhibited by these factors, which include regulatory T cells, myeloid-derived suppressor cells and immunosuppressive molecules (127,128). Thus, it is imperative to overcome these immunosuppressive factors to improve the survival rate and antitumor activity of iPSC-NK cells in the tumor microenvironment.

Tumor heterogeneity poses a significant challenge for iPSC-NK cells cell therapy. Genetic and epigenetic heterogeneity among tumor cells drive the emergence of diverse cellular subpopulations within tumors, resulting in varied responses to treatment and uncertainty in therapeutic efficacy. Studies have indicated that NK cells do not persist well in vivo, which limits the durability of their response against tumors (129131). Therefore, enhancing the persistence of iPSC-NK cells is crucial for their therapeutic efficacy in vivo.

In summary, although iPSC-NK cells have the potential to recognize and kill tumor cells, several challenges must be addressed for their clinical application: Enhancing in vivo persistence, improving tumor targeting, overcoming the effects of the tumor microenvironment, optimizing differentiation strategies, achieving scalable production and quality control and managing immunogenicity and toxicity issues. Improving the specificity and targeting efficiency in the complex tumor microenvironment will aid in advancing the development and application of iPSC-NK cells therapies.

Discussion and future perspectives

To date, only Fate Therapeutics has completed a phase I clinical trial using iPSC-NK cells to treat cancer (107). Partial clinical trial data indicate that both genetically modified and unmodified iPSC-NK cells have demonstrated good safety and tolerability, with no severe adverse events related to NK cells (58,73,109). It has been reported that unmodified iPSC-NK cells show therapeutic effects against various types of tumors, especially in solid tumors (62). Meanwhile, genetically modified iPSC-NK cells have displayed favorable overall response rates and complete response rates in treating relapsed or refractory lymphomas (110). The development of CAR-iPSC-NK cells has brought new hope to the field of cancer therapy (6,88,132).

Despite these promising results, potential risks associated with iPSC-NK cells remain. Hypoxia and the accumulation of metabolites are two factors that may affect the metabolic adaptability and function of CAR-NK cells. For example, the activation receptors of CAR-NK cells may be inhibited in hypoxic environments, leading to a reduction in their cytotoxicity. Additionally, NK cells can enhance their utilization of glucose to promote glycolysis and mitochondrial oxidative phosphorylation. This metabolic adaptation provides sufficient energy and biosynthetic precursors, thereby supporting their proliferation and cytotoxic functions. (133,134).

The rapid utilization of glucose by tumor cells may also induce metabolic reprogramming in NK cells, thereby affecting their effector functions (123). The specificity and durability of CAR-iPSC derived NK cells have been further enhanced by recent advances in CRISPR-Cas9-based gene editing technology. For example, Shankar et al (135) used non-viral CRISPR gene editing technology on iPSC-NK cells to successfully insert CAR to GD2 while knocking out the KLRC1 gene. This modification markedly enhanced the ability to kill solid tumors. The improvements enhance the NK cells to proliferate by prolonging their survival in vivo and alleviating inhibitory signals. This improvement may be partly attributed to modifications in CAR that influence cellular metabolism. Therefore, further in-depth research is needed to explore strategies for overcoming the immunosuppressive effects of metabolites through CAR-iPSC-NK cells.

In the context of combination therapies, integrating iPSC-NK cells with monoclonal antibodies targeting tumor antigens may further enhance tumor recognition and eradication. Existing studies show that iPSC-NK cells and PD-1/PD-L1 inhibitors can synergistically enhance anti-tumor efficacy by overcoming immunosuppression in the tumor microenvironment, ultimately improving anti-tumor immune responses (136,137). Currently, clinical trials employing iPSC-NK cells in combination with monoclonal antibodies mainly include FT596 and CD19t-haNK (NCT06334991). CD19t-haNK is a clinical trial evaluating CAR-NK as a monotherapy or in combination with rituximab for treating relapsed/refractory CD19 and CD20 B-cell non-Hodgkin lymphoma (138). The first patients were successfully dosed in October 2024, with additional data expected in subsequent phases.

As reprogramming factors such as Oct3/4, Sox2, Klf4 and c-Myc are closely associated with tumorigenesis, their use in iPSC generation raises concerns. Notably, c-Myc mutations are frequent in human cancers, further highlighting the need for caution in iPSC-based therapies. To fully unlock the potential of iPSC-NK cells therapies in oncology, rigorous safety measures and technological advancements are critical for addressing these risks (139). Therefore, meticulous control of the iPSC cell amplification process is necessary. This includes selecting iPSC lines that have already demonstrated stability and safety in preclinical studies and employing gene-editing techniques to repair or correct potential mutation-causing sites to enhance iPSC stability. For example, to replace oncogenic factors, Ding et al (140) employed transient mRNA delivery or small molecules.

Meanwhile, single-cell RNA sequencing technology can markedly contribute to resolving the heterogeneity during the differentiation process of iPSCs. By identifying functional subsets of NK cells, it can optimize the differentiation strategy of iPSC-NK cells (42,117). This technology has been used to analyze gene expression differences in iPSC-NK cells within the tumor microenvironment, shedding light on their interaction mechanisms with tumor cells, immunosuppressive cells and stromal cells. Additionally, single-cell sequencing has the potential to identify new therapeutic targets, thereby advancing iPSC-NK cells therapy in cancer treatment and immunotherapy (141). Using single-cell sequencing technology help detect abnormal genomic mutations, thereby reducing potential iPSC-related risks.

IPSC-NK cells are emerging as a novel and promising cell therapy tool in multiple ongoing clinical studies for solid tumor. They have demonstrated safety and potential efficacy in several reported clinical studies. However, some challenges remain in the clinical application of iPSC-NK cells, such as tumor microenvironment, differentiation strategies and quality control. Thus, more preclinical and clinical studies are required to push the iPSC-NK therapy into clinical applications. The establishment of allogeneic iPSC-NK cells as a next-generation immunotherapy, particularly in oncology, will be strongly supported by overcoming these hurdles.

Acknowledgements

Not applicable.

Funding

Funding: No funding was received.

Availability of data and materials

Not applicable.

Authors' contributions

XW, CS, YL, NW, KX, QQ and ZX wrote and revised the manuscript. ZX and CS designed and supervised the study. YL and NW reviewed the references. QQ and KX provided supervision, reviewing and editing of the final manuscript. Data authentication is not applicable. All authors read and approved the final manuscript.

Ethics approval and consent to participate

Not applicable.

Patient consent for publication

Not applicable.

Competing interests

Not applicable.

Glossary

Abbreviations

Abbreviations:

NK

natural killer

IPSC

induced pluripotent stem cell

CAR

chimeric antigen receptor

CR

complete response

CRS

cytokine release syndrome

ADCC

antibody-dependent cellular cytotoxicity

DCs

dendritic cells

IFN

interferon

TNF

tumor necrosis factor

XCL

recombinant chemokine C-motif ligand

CCR

C-C chemokine receptor

MHC

major histocompatibility complex

HLA

human leukocyte antigen

NCR

natural cytotoxicity receptor

KIR

killer cell immunoglobulin-like receptor

EB

spin embryoid body

HPCs

hematopoietic progenitor cells

KIRs

killer immunoglobulin-like receptors

PD-1

anti- programmed death-1

PD-L1

Programmed cell death ligand 1

ADAM17

a disintegrin and metalloproteinase

CD16 non-cleavable

hnCD16

TIGIT

T cell immune receptor with immunoglobulin and ITIM domains

B2M

β-2 macroglobulin

scFv

single-chain variable fragment

MM

Multiple myeloma

References

1 

Bald T, Krummel MF, Smyth MJ and Barry KC: The NK cell-cancer cycle: Advances and new challenges in NK cell-based immunotherapies. Nat Immunol. 21:835–847. 2020. View Article : Google Scholar : PubMed/NCBI

2 

Wang D, Sun Z, Zhu X, Zheng X, Zhou Y, Lu Y, Yan P, Wang H, Liu H, Jin J, et al: GARP-mediated active TGF-beta1 induces bone marrow NK cell dysfunction in AML patients with early relapse post-allo-HSCT. Blood. 140:2788–2804. 2022. View Article : Google Scholar : PubMed/NCBI

3 

Terren I, Orrantia A, Vitalle J, Astarloa-Pando G, Zenarruzabeitia O and Borrego F: Modulating NK cell metabolism for cancer immunotherapy. Semin Hematol. 57:213–224. 2020. View Article : Google Scholar : PubMed/NCBI

4 

Hernani R, Benzaquen A and Solano C: Toxicities following CAR-T therapy for hematological malignancies. Cancer Treat Rev. 111:1024792022. View Article : Google Scholar : PubMed/NCBI

5 

Mansour AG, Teng KY, Li Z, Zhu Z, Chen H, Tian L, Ali A, Zhang J, Lu T, Ma S, et al: Off-the-shelf CAR-engineered natural killer cells targeting FLT3 enhance killing of acute myeloid leukemia. Blood Adv. 7:6225–6239. 2023. View Article : Google Scholar : PubMed/NCBI

6 

Wu X and Matosevic S: Gene-edited and CAR-NK cells: Opportunities and challenges with engineering of NK cells for immunotherapy. Mol Ther Oncolytics. 27:224–238. 2022. View Article : Google Scholar : PubMed/NCBI

7 

Raneros AB, Lopez-Larrea C and Suarez-Alvarez B: Acute myeloid leukemia and NK cells: Two warriors confront each other. Oncoimmunology. 8:e15396172019. View Article : Google Scholar : PubMed/NCBI

8 

Woan KV, Kim H, Bjordahl R, Davis ZB, Gaidarova S, Goulding J, Hancock B, Mahmood S, Abujarour R, Wang H, et al: Harnessing features of adaptive NK cells to generate iPSC-derived NK cells for enhanced immunotherapy. Cell Stem Cell. 28:2062–2075.e2065. 2021. View Article : Google Scholar : PubMed/NCBI

9 

Li Y, Hermanson DL, Moriarity BS and Kaufman DS: Human iPSC-Derived natural killer cells engineered with chimeric antigen receptors enhance anti-tumor activity. Cell Stem Cell. 23:181–192.e185. 2018. View Article : Google Scholar : PubMed/NCBI

10 

Lin X, Sun Y, Dong X, Liu Z, Sugimura R and Xie G: IPSC-derived CAR-NK cells for cancer immunotherapy. Biomed Pharmacother. 165:1151232023. View Article : Google Scholar : PubMed/NCBI

11 

Zhu H, Blum RH, Bernareggi D, Ask EH, Wu Z, Hoel HJ, Meng Z, Wu C, Guan KL, Malmberg KJ and Kaufman DS: Metabolic reprograming via deletion of CISH in human iPSC-derived NK cells promotes in vivo persistence and enhances anti-tumor activity. Cell Stem Cell. 27:224–237.e226. 2020. View Article : Google Scholar : PubMed/NCBI

12 

Farahzadi R, Valipour B, Anakok OF, Fathi E and Montazersaheb S: The effects of encapsulation on NK cell differentiation potency of C-kit+ hematopoietic stem cells via identifying cytokine profiles. Transpl Immunol. 77:1017972023. View Article : Google Scholar : PubMed/NCBI

13 

Mace EM: Human natural killer cells: Form, function, and development. J Allergy Clin Immunol. 151:371–385. 2023. View Article : Google Scholar : PubMed/NCBI

14 

Galat Y, Du Y, Perepitchka M, Li XN, Balyasnikova IV, Tse WT, Dambaeva S, Schneiderman S, Iannaccone PM, Becher O, et al: In vitro vascular differentiation system efficiently produces natural killer cells for cancer immunotherapies. Oncoimmunology. 12:22406702023. View Article : Google Scholar : PubMed/NCBI

15 

Vacca P, Vitale C, Montaldo E, Conte R, Cantoni C, Fulcheri E, Darretta V, Moretta L and Mingari MC: CD34+ hematopoietic precursors are present in human decidua and differentiate into natural killer cells upon interaction with stromal cells. Proc Natl Acad Sci USA. 108:2402–2407. 2011. View Article : Google Scholar : PubMed/NCBI

16 

López-Botet M, De Maria A, Muntasell A, Della Chiesa M and Vilches C: Adaptive NK cell response to human cytomegalovirus: Facts and open issues. Semin Immunol. 65:1017062023. View Article : Google Scholar : PubMed/NCBI

17 

Panjwani MK, Grassmann S, Sottile R, Le Luduec JB, Kontopoulos T, van der Ploeg K, Sun JC and Hsu KC: Single-cell profiling aligns CD56bright and cytomegalovirus-induced adaptive natural killer cells to a naïve-memory relationship. Front Immunol. 15:14994922024. View Article : Google Scholar : PubMed/NCBI

18 

Liu M, Liu J, Zhang X, Xiao Y, Jiang G and Huang X: Activation status of CD56 dim natural killer cells is associated with disease activity of patients with systemic lupus erythematosus. Clin Rheumatol. 40:1103–1112. 2021. View Article : Google Scholar : PubMed/NCBI

19 

Oboshi W, Watanabe T, Matsuyama Y, Kobara A, Yukimasa N, Ueno I, Aki K, Tada T and Hosoi E: The influence of NK cell-mediated ADCC: Structure and expression of the CD16 molecule differ among FcγRIIIa-V158F genotypes in healthy Japanese subjects. Hum Immunol. 77:165–171. 2016. View Article : Google Scholar : PubMed/NCBI

20 

De Federicis D, Capuano C, Ciuti D, Molfetta R, Galandrini R and Palmieri G: Nutrient transporter pattern in CD56dim NK cells: CD16 (FcγRIIIA)-dependent modulation and association with memory NK cell functional profile. Front Immunol. 15:14777762024. View Article : Google Scholar : PubMed/NCBI

21 

Wagner JA, Rosario M, Romee R, Berrien-Elliott MM, Schneider SE, Leong JW, Sullivan RP, Jewell BA, Becker-Hapak M, Schappe T, et al: CD56 bright NK cells exhibit potent antitumor responses following IL-15 priming. J Clin Invest. 127:4042–4058. 2017. View Article : Google Scholar : PubMed/NCBI

22 

Enomoto Y, Li P, Jenkins LM, Anastasakis D, Lyons GC, Hafner M and Leonard WJ: Cytokine-enhanced cytolytic activity of exosomes from NK Cells. Cancer Gene Ther. 29:734–749. 2022. View Article : Google Scholar : PubMed/NCBI

23 

Lachota M, Zielniok K, Palacios D, Kanaya M, Penna L, Hoel HJ, Wiiger MT, Kveberg L, Hautz W, Zagożdżon R and Malmberg KJ: Mapping the chemotactic landscape in NK cells reveals subset-specific synergistic migratory responses to dual chemokine receptor ligation. EBioMedicine. 96:1048112023. View Article : Google Scholar : PubMed/NCBI

24 

Toffoli E, van Vliet A, Forbes C, Arns AJ, Verheul HWM, Tuynman J, van der Vliet HJ, Spanholtz J and de Gruijl TD: Allogeneic NK cells induce the in vitro activation of monocyte-derived and conventional type-2 dendritic cells and trigger an inflammatory response under cancer-associated conditions. Clin Exp Immunol. 216:159–171. 2024. View Article : Google Scholar : PubMed/NCBI

25 

Rodriguez-Mogeda C, van Ansenwoude CM, van der Molen L, Strijbis EM, Mebius RE and de Vries HE: The role of CD56bright NK cells in neurodegenerative disorders. J Neuroinflammation. 21:482024. View Article : Google Scholar : PubMed/NCBI

26 

van de Donk N, Richardson PG and Malavasi F: CD38 antibodies in multiple myeloma: Back to the future. Blood. 131:13–29. 2018. View Article : Google Scholar : PubMed/NCBI

27 

Wagner JA and Fehniger TA: Human adaptive natural killer cells: Beyond NKG2C. Trends Immunol. 37:351–353. 2016. View Article : Google Scholar : PubMed/NCBI

28 

Zappa E, Vitali A, Anders K, Molenaar JJ, Wienke J and Künkele A: Adoptive cell therapy in pediatric extracranial solid tumors: Current approaches and future challenges. Eur J Cancer. 194:1133472023. View Article : Google Scholar : PubMed/NCBI

29 

Taylor BC and Balko JM: Mechanisms of MHC-I downregulation and role in immunotherapy response. Front Immunol. 13:8448662022. View Article : Google Scholar : PubMed/NCBI

30 

Shreeve N, Depierreux D, Hawkes D, Traherne JA, Sovio U, Huhn O, Jayaraman J, Horowitz A, Ghadially H, Perry JRB, et al: The CD94/NKG2A inhibitory receptor educates uterine NK cells to optimize pregnancy outcomes in humans and mice. Immunity. 54:1231–1244.e1234. 2021. View Article : Google Scholar : PubMed/NCBI

31 

Pollock NR, Harrison GF and Norman PJ: Immunogenomics of killer cell immunoglobulin-like receptor (KIR) and HLA class I: Coevolution and consequences for human health. J Allergy Clin Immunol Pract. 10:1763–1775. 2022. View Article : Google Scholar : PubMed/NCBI

32 

Rascle P, Woolley G, Jost S, Manickam C and Reeves RK: NK cell education: Physiological and pathological influences. Front Immunol. 14:10871552023. View Article : Google Scholar : PubMed/NCBI

33 

Neo SY, Jing X, Tong L, Tong D, Gao J, Chen Z, De Los Santos MC, Burduli N, De Souza Ferreira S, Wagner AK, et al: Tumor MHC class I expression alters cancer-associated myelopoiesis driven by host NK cells. J Immunother Cancer. 10:e0053082022. View Article : Google Scholar : PubMed/NCBI

34 

Chen X, Lu Q, Zhou H, Liu J, Nadorp B, Lasry A, Sun Z, Lai B, Rona G, Zhang J, et al: A membrane-associated MHC-I inhibitory axis for cancer immune evasion. Cell. 186:3903–3920.e3921. 2023. View Article : Google Scholar : PubMed/NCBI

35 

Toyoda H, Kuramasu A, Hosonuma M, Murayama M, Narikawa Y, Isobe J, Baba Y, Tajima K, Funayama E, Shida M, et al: MHC class I polypeptide-related sequence B shedding modulates pancreatic tumor immunity via the activation of NKG2DLow T cells. Sci Rep. 14:234012024. View Article : Google Scholar : PubMed/NCBI

36 

Harkus U, Wankell M, Palamuthusingam P, McFarlane C and Hebbard L: Immune checkpoint inhibitors in HCC: Cellular, molecular and systemic data. Semin Cancer Biol. 86:799–815. 2022. View Article : Google Scholar : PubMed/NCBI

37 

Eugene J, Jouand N, Ducoin K, Dansette D, Oger R, Deleine C, Leveque E, Meurette G, Podevin J, Matysiak T, et al: The inhibitory receptor CD94/NKG2A on CD8(+) tumor-infiltrating lymphocytes in colorectal cancer: A promising new druggable immune checkpoint in the context of HLAE/β2m overexpression. Mod Pathol. 33:468–482. 2020. View Article : Google Scholar : PubMed/NCBI

38 

Sun C, Xu J, Huang Q, Huang M, Wen H, Zhang C, Wang J, Song J, Zheng M, Sun H, et al: High NKG2A expression contributes to NK cell exhaustion and predicts a poor prognosis of patients with liver cancer. Oncoimmunology. 6:e12645622017. View Article : Google Scholar : PubMed/NCBI

39 

Mahgoub S, Abosalem H, Emara M, Kotb N, Maged A and Soror S: Restoring NK cells functionality via cytokine activation enhances cetuximab-mediated NK-cell ADCC: A promising therapeutic tool for HCC patients. Mol Immunol. 137:221–227. 2021. View Article : Google Scholar : PubMed/NCBI

40 

Hò GGT, Celik AA, Huyton T, Hiemisch W, Blasczyk R, Simper GS and Bade-Doeding C: NKG2A/CD94 is a new immune receptor for HLA-G and distinguishes amino acid differences in the HLA-G heavy chain. Int J Mol Sci. 21:43622020. View Article : Google Scholar : PubMed/NCBI

41 

Laskowski TJ, Biederstadt A and Rezvani K: Natural killer cells in antitumour adoptive cell immunotherapy. Nat Rev Cancer. 22:557–575. 2022. View Article : Google Scholar : PubMed/NCBI

42 

Bibby JA, Agarwal D, Freiwald T, Kunz N, Merle NS, West EE, Singh P, Larochelle A, Chinian F, Mukherjee S, et al: Systematic single-cell pathway analysis to characterize early T cell activation. Cell Rep. 41:1116972022. View Article : Google Scholar : PubMed/NCBI

43 

Wong JKM, Dolcetti R, Rhee H, Simpson F and Souza-Fonseca-Guimaraes F: Weaponizing natural killer cells for solid cancer immunotherapy. Trends Cancer. 9:111–121. 2023. View Article : Google Scholar : PubMed/NCBI

44 

Dixon KJ, Wu J and Walcheck B: Engineering anti-tumor monoclonal antibodies and Fc receptors to enhance ADCC by human NK cells. Cancers (Basel). 13:3122021. View Article : Google Scholar : PubMed/NCBI

45 

Kim IY, Kim HY, Song HW, Park JO, Choi YH and Choi E: Functional enhancement of exosomes derived from NK cells by IL-15 and IL-21 synergy against hepatocellular carcinoma cells: The cytotoxicity and apoptosis in vitro study. Heliyon. 9:e169622023. View Article : Google Scholar : PubMed/NCBI

46 

Bottino C, Castriconi R, Moretta L and Moretta A: Cellular ligands of activating NK receptors. Trends Immunol. 26:221–226. 2005. View Article : Google Scholar : PubMed/NCBI

47 

Zhang R, Liu Q, Zhou S, He H, Zhao M and Ma W: Engineering CAR-NK cells targeting CD33 with concomitant extracellular secretion of anti-CD16 antibody revealed superior antitumor effects toward myeloid leukemia. Cancer Lett. 558:2161032023. View Article : Google Scholar : PubMed/NCBI

48 

Raulet DH: Roles of the NKG2D immunoreceptor and its ligands. Nat Rev Immunol. 3:781–790. 2003. View Article : Google Scholar : PubMed/NCBI

49 

Kohlhapp FJ, O'Sullivan JA, Moore TV, Zloza A and Guevara-Patino JA: NKG2D signaling shifts the balance of CD8 T cells from single cytokine- to polycytokine-producing effector cells. Mol Immunol. 155:1–6. 2023. View Article : Google Scholar : PubMed/NCBI

50 

Corvino D, Kumar A and Bald T: Plasticity of NK cells in cancer. Front Immunol. 13:8883132022. View Article : Google Scholar : PubMed/NCBI

51 

Asl KD, Rafat A, Mazloumi Z, Valipour B, Movassaghpour A, Talebi M, Mahdavi M, Nasrabadi HT and Charoudeh HN: Cord blood stem cell-generated KIR(+)NK cells effectively target leukemia cell lines. Hum Immunol. 84:98–105. 2023. View Article : Google Scholar : PubMed/NCBI

52 

Klingemann H: The NK-92 cell line-30 years later: Its impact on natural killer cell research and treatment of cancer. Cytotherapy. 25:451–457. 2023. View Article : Google Scholar : PubMed/NCBI

53 

García Aponte OF, Kozma B, Egger D, Kasper C and Herwig C: Kinetics of NK-92 growth and functionality in pseudo-static cultures. Biochemical Engineering J. 196:1089292023. View Article : Google Scholar

54 

Clara JA, Levy ER, Reger R, Barisic S, Chen L, Cherkasova E, Chakraborty M, Allan DSJ and Childs R: High-affinity CD16 integration into a CRISPR/Cas9-edited CD38 locus augments CD38-directed antitumor activity of primary human natural killer cells. J Immunother Cancer. 10:e0038042022. View Article : Google Scholar : PubMed/NCBI

55 

Takahashi K and Yamanaka S: Induction of pluripotent stem cells from mouse embryonic and adult fibroblast cultures by defined factors. Cell. 126:663–676. 2006. View Article : Google Scholar : PubMed/NCBI

56 

Crow D: Could iPSCs enable ‘Off-the-Shelf’ cell therapy? Cell. 177:1667–1669. 2019. View Article : Google Scholar : PubMed/NCBI

57 

Naama M and Buganim Y: Human trophoblast stem cell-state acquisition from pluripotent stem cells and somatic cells. Curr Opin Genet Dev. 81:1020842023. View Article : Google Scholar : PubMed/NCBI

58 

Goodridge JP, Mahmood S, Zhu H, Gaidarova S, Blum R, Bjordahl R, Cichocki F, Chu H, Bonello G, Lee T, et al: FT596: Translation of first-of-kind multi-antigen targeted off-the-shelf CAR-NK cell with engineered persistence for the treatment of B cell malignancies. Blood. 134:3012019. View Article : Google Scholar

59 

Valamehr B, Robinson M, Abujarour R, Rezner B, Vranceanu F, Le T, Medcalf A, Lee TT, Fitch M, Robbins D and Flynn P: Platform for induction and maintenance of transgene-free hiPSCs resembling ground state pluripotent stem cells. Stem Cell Reports. 2:366–381. 2014. View Article : Google Scholar : PubMed/NCBI

60 

Kiran S, Xue Y, Sarker DB, Li Y and Sang QXA: Feeder-free differentiation of human iPSCs into natural killer cells with cytotoxic potential against malignant brain rhabdoid tumor cells. Bioact Mater. 36:301–316. 2024.PubMed/NCBI

61 

Lv Y, Rao Z, Liu L, Jia J, Wu C, Xu J, Du Y, Liu Y, Liu B, Shi J, et al: The efficient generation of functional human hepatocytes from chemically induced pluripotent stem cells. Cell Prolif. 57:e135402024. View Article : Google Scholar : PubMed/NCBI

62 

Lupo KB, Moon JI, Chambers AM and Matosevic S: Differentiation of natural killer cells from induced pluripotent stem cells under defined, serum- and feeder-free conditions. Cytotherapy. 23:939–952. 2021. View Article : Google Scholar : PubMed/NCBI

63 

Denman CJ, Senyukov VV, Somanchi SS, Phatarpekar PV, Kopp LM, Johnson JL, Singh H, Hurton L, Maiti SN, Huls MH, et al: Membrane-bound IL-21 promotes sustained ex vivo proliferation of human natural killer cells. PLoS One. 7:e302642012. View Article : Google Scholar : PubMed/NCBI

64 

Knorr DA, Ni Z, Hermanson D, Hexum MK, Bendzick L, Cooper LJ, Lee DA and Kaufman DS: Clinical-scale derivation of natural killer cells from human pluripotent stem cells for cancer therapy. Stem Cells Transl Med. 2:274–283. 2013. View Article : Google Scholar : PubMed/NCBI

65 

Qiao W, Dong P, Chen H and Zhang J: Advances in induced pluripotent stem cell-derived natural killer cell therapy. Cells. 13:19762024. View Article : Google Scholar : PubMed/NCBI

66 

Zhang L, Weiskittel TM, Zhu Y, Xue D, Zhang H, Shen Y, Yu H, Li J, Hou L, Guo H, et al: Comparative dissection of transcriptional landscapes of human iPSC-NK differentiation and NK cell development. Life Med. 3:lnae0322024. View Article : Google Scholar : PubMed/NCBI

67 

Thangaraj JL, Coffey M, Lopez E and Kaufman DS: Disruption of TGF-β signaling pathway is required to mediate effective killing of hepatocellular carcinoma by human iPSC-derived NK cells. Cell Stem Cell. 31:1327–1343.e1325. 2024. View Article : Google Scholar : PubMed/NCBI

68 

Qin Y, Cui Q, Sun G, Chao J, Wang C, Chen X, Ye P, Zhou T, Jeyachandran AV, Sun O, et al: Developing enhanced immunotherapy using NKG2A knockout human pluripotent stem cell-derived NK cells. Cell Rep. 43:1148672024. View Article : Google Scholar : PubMed/NCBI

69 

Goldenson BH, Hor P and Kaufman DS: iPSC-derived natural killer cell therapies-expansion and targeting. Front Immunol. 13:8411072022. View Article : Google Scholar : PubMed/NCBI

70 

Bernareggi D, Gonsalves C, Schabla M, Gárate-Carrillo A, El-Kalay M, Kaufman DS, Hollingsworth R and Zhu H: 336 A novel method for efficient cGMP production of natural killer cells from clonal master induced pluripotent stem cells for next generation, off-the-shelf cancer immunotherapy. Regular and Young Investigator Award Abstracts. A354. 2022. View Article : Google Scholar

71 

Maddineni S, Silberstein JL and Sunwoo JB: Emerging NK cell therapies for cancer and the promise of next generation engineering of iPSC-derived NK cells. J Immunother Cancer. 10:e0046932022. View Article : Google Scholar : PubMed/NCBI

72 

Patel M, Park D, Tarantolo S, Dowlati A, Olson D, Kaneko Y, Tang M, Soukharev S, Takizawa M, Okada Y, et al: 754 A phase 1/2 study of ASP1570 in participants with locally advanced or metastatic solid tumors who have progressed on, or are ineligible for, all available standard therapies. Regular and Young Investigator Award Abstracts. A786. 2022. View Article : Google Scholar

73 

Bachanova V, Ghobadi A, Patel K, Park JH, Flinn IW, Shah P, Wong C, Bickers C, Szabo P, Wong L, et al: Safety and efficacy of FT596, a first-in-class, multi-antigen targeted, off-the-shelf, iPSC-derived CD19 CAR NK cell therapy in relapsed/refractory B-Cell lymphoma. Blood. 138:823. 2021. View Article : Google Scholar

74 

Tang SY, Zha S, Du Z, Zeng J, Zhu D, Luo Y and Wang S: Targeted integration of EpCAM-specific CAR in human induced pluripotent stem cells and their differentiation into NK cells. Stem Cell Res Ther. 12:5802021. View Article : Google Scholar : PubMed/NCBI

75 

Ma S, Caligiuri MA and Yu J: Harnessing IL-15 signaling to potentiate NK cell-mediated cancer immunotherapy. Trends Immunol. 43:833–847. 2022. View Article : Google Scholar : PubMed/NCBI

76 

Vahidi S, Touchaei AZ and Samadani AA: IL-15 as a key regulator in NK cell-mediated immunotherapy for cancer: From bench to bedside. Int Immunopharmacol. 133:1121562024. View Article : Google Scholar : PubMed/NCBI

77 

Seo IH, Eun HS, Kim JK, Lee H, Jeong S, Choi SJ, Lee J, Lee BS, Kim SH, Rou WS, et al: IL-15 enhances CCR5-mediated migration of memory CD8(+) T cells by upregulating CCR5 expression in the absence of TCR stimulation. Cell Rep. 36:1094382021. View Article : Google Scholar : PubMed/NCBI

78 

Valipour B, Abedelahi A, Naderali E, Velaei K, Movassaghpour A, Talebi M, Montazersaheb S, Karimipour M, Darabi M, Chavoshi H, et al: Cord blood stem cell derived CD16(+) NK cells eradicated acute lymphoblastic leukemia cells using with anti-CD47 antibody. Life Sci. 242:1172232020. View Article : Google Scholar : PubMed/NCBI

79 

Quatrini L, Vacca P, Tumino N, Besi F, Di Pace AL, Scordamaglia F, Martini S, Munari E, Mingari MC, Ugolini S and Moretta L: Glucocorticoids and the cytokines IL-12, IL-15, and IL-18 present in the tumor microenvironment induce PD-1 expression on human natural killer cells. J Allergy Clin Immunol. 147:349–360. 2021. View Article : Google Scholar : PubMed/NCBI

80 

Chen AP, Gao P, Lin L, Ashok P, He H, Ma C, Zou DL, Allain V, Boyne A, Juillerat A, et al: An improved approach to generate IL-15+/+/TGFβR2−/− iPSC-derived natural killer cells using TALEN. Cell Rep Methods. 4:1008572024. View Article : Google Scholar : PubMed/NCBI

81 

Fares J, Davis ZB, Rechberger JS, Toll SA, Schwartz JD, Daniels DJ, Miller JS and Khatua S: Advances in NK cell therapy for brain tumors. NPJ Precis Oncol. 7:172023. View Article : Google Scholar : PubMed/NCBI

82 

Kim J, Phan MTT, Hwang I, Park J and Cho D: Comparison of the different anti-CD16 antibody clones in the activation and expansion of peripheral blood NK cells. Sci Rep. 13:94932023. View Article : Google Scholar : PubMed/NCBI

83 

Capuano C, Pighi C, Battella S, De Federicis D, Galandrini R and Palmieri G: Harnessing CD16-mediated NK cell functions to enhance therapeutic efficacy of tumor-targeting mAbs. Cancers (Basel). 13:25002021. View Article : Google Scholar : PubMed/NCBI

84 

Romee R, Foley B, Lenvik T, Wang Y, Zhang B, Ankarlo D, Luo X, Cooley S, Verneris M, Walcheck B and Miller J: NK cell CD16 surface expression and function is regulated by a disintegrin and metalloprotease-17 (ADAM17). Blood. 121:3599–3608. 2013. View Article : Google Scholar : PubMed/NCBI

85 

Wang Y, Wu J, Newton R, Bahaie NS, Long C and Walcheck B: ADAM17 cleaves CD16b (FcγRIIIb) in human neutrophils. Biochim Biophys Acta. 1833:680–685. 2013. View Article : Google Scholar : PubMed/NCBI

86 

Meng F, Zhang S, Xie J, Zhou Y, Wu Q, Lu B, Zhou S, Zhao X and Li Y: Leveraging CD16 fusion receptors to remodel the immune response for enhancing anti-tumor immunotherapy in iPSC-derived NK cells. J Hematol Oncol. 16:622023. View Article : Google Scholar : PubMed/NCBI

87 

Yamamoto K, Blum R and Kaufman DS: ADAM17-deficient pluripotent stem cell-derived natural killer cells possess improved antibody-dependent cellular cytotoxicity and antitumor activity. Blood. 136:22020. View Article : Google Scholar

88 

Biederstädt A and Rezvani K: Engineering the next generation of CAR-NK immunotherapies. Int J Hematol. 114:554–571. 2021. View Article : Google Scholar : PubMed/NCBI

89 

Gao L, Du X, Li J and Qin FXF: Evolving roles of CD38 metabolism in solid tumour microenvironment. Br J Cancer. 128:492–504. 2023. View Article : Google Scholar : PubMed/NCBI

90 

Stikvoort A, van der Schans J, Sarkar S, Poels R, Ruiter R, Naik J, Yuan H, de Bruijn JD, van de Donk NWCJ, Zweegman S, et al: CD38-specific chimeric antigen receptor expressing natural killer KHYG-1 cells: A proof of concept for an ‘Off the Shelf’ therapy for multiple myeloma. Hemasphere. 5:e5962021. View Article : Google Scholar : PubMed/NCBI

91 

Seiffert M: TIGIT: An immune checkpoint beyond T cells in chronic lymphocytic leukemia. Haematologica. 108:1979–1981. 2023. View Article : Google Scholar : PubMed/NCBI

92 

Neo SY, Yang Y, Record J, Ma R, Chen X, Chen Z, Tobin NP, Blake E, Seitz C, Thomas R, et al: CD73 immune checkpoint defines regulatory NK cells within the tumor microenvironment. J Clin Invest. 130:1185–1198. 2020. View Article : Google Scholar : PubMed/NCBI

93 

Lupo KB, Yao X, Borde S, Wang J, Torregrosa-Allen S, Elzey BD, Utturkar S, Lanman NA, McIntosh M and Matosevic S: synNotch-programmed iPSC-derived NK cells usurp TIGIT and CD73 activities for glioblastoma therapy. Nat Commun. 15:19092024. View Article : Google Scholar : PubMed/NCBI

94 

Reiser J, Chan SR, Mathavan K, Sillitti D, Mottershead C, Mattson B, Pache M, Gutierrez A, Scoon W, Zhu Y, et al: FT555: Off-the-Shelf CAR-NK cell therapy co-targeting GPRC5D and CD38 for the treatment of multiple myeloma. Blood. 140:4560–4561. 2022. View Article : Google Scholar

95 

Vahidian F, Khosroshahi LM, Akbarzadeh M, Jahanban-Esfahlan A, Baghbanzadeh A, Ali-Hassanzadeh M and Safarzadeh E: The tricks for fighting against cancer using CAR NK cells: A review. Mol Cell Probes. 63:1018172022. View Article : Google Scholar : PubMed/NCBI

96 

Marofi F, Abdul-Rasheed OF, Rahman HS, Budi HS, Jalil AT, Yumashev AV, Hassanzadeh A, Yazdanifar M, Motavalli R, Chartrand MS, et al: CAR-NK cell in cancer immunotherapy; A promising frontier. Cancer Sci. 112:3427–3436. 2021. View Article : Google Scholar : PubMed/NCBI

97 

Cichocki F, Bjordahl R, Gaidarova S, Mahmood S, Abujarour R, Wang H, Tuininga K, Felices M, Davis ZB, Bendzick L, et al: iPSC-derived NK cells maintain high cytotoxicity and enhance in vivo tumor control in concert with T cells and anti-PD-1 therapy. Sci Transl Med. 12:eaaz56182020. View Article : Google Scholar : PubMed/NCBI

98 

Karagiannis P and Kim SI: iPSC-derived natural killer cells for cancer immunotherapy. Mol Cells. 44:541–548. 2021. View Article : Google Scholar : PubMed/NCBI

99 

Karvouni M, Vidal-Manrique M, Susek KH, Hussain A, Gilljam M, Zhang Y, Gray JD, Lund J, Kaufmann G, Ljunggren HG, et al: Challenges in alphaCD38-chimeric antigen receptor (CAR)-expressing natural killer (NK) cell-based immunotherapy in multiple myeloma: Harnessing the CD38dim phenotype of cytokine-stimulated NK cells as a strategy to prevent fratricide. Cytotherapy. 25:763–772. 2023. View Article : Google Scholar : PubMed/NCBI

100 

Bjordahl R, Gaidarova S, Goodridge JP, Mahmood S, Bonello G, Robinson M, Ruller C, Pribadi M, Lee T, Abujarour R, et al: FT576: A novel multiplexed engineered off-the-shelf natural killer cell immunotherapy for the dual-targeting of CD38 and Bcma for the treatment of multiple myeloma. Blood. 134:32142019. View Article : Google Scholar

101 

Goodridge JP, Bjordahl R, Mahmood S, Reiser J, Gaidarova S, Blum R, Cichocki F, Chu H, Bonello G, Lee T, et al: FT576: Multi-Specific Off-the-shelf CAR-NK cell therapy engineered for enhanced persistence, avoidance of self-fratricide and optimized mab combination therapy to prevent antigenic escape and elicit a deep and durable response in multiple myeloma. Blood. 136:4–5. 2020. View Article : Google Scholar

102 

Dhakal B, Berdeja JG, Gregory T, Ly T, Bickers C, Zong X, Wong L, Goodridge JP, Cooley S, Valamehr B, et al: Interim phase I clinical data of FT576 as monotherapy and in combination with daratumumab in subjects with relapsed/refractory multiple myeloma. Blood. 140:4586–4587. 2022. View Article : Google Scholar

103 

Wang L, Wang Y, He X, Mo Z, Zhao M, Liang X, Hu K, Wang K, Yue Y, Mo G, et al: CD70-targeted iPSC-derived CAR-NK cells display potent function against tumors and alloreactive T cells. Cell Rep Med. 6:1018892025. View Article : Google Scholar : PubMed/NCBI

104 

Yu M, Mansour AG, Teng KY, Sun G, Shi Y and Caligiuri MA: iPSC-derived natural killer cells expressing EGFR-CAR against glioblastoma. Cancer Research. 80:33132020. View Article : Google Scholar

105 

Wang Y, Wang L, Shao M, He X, Yue Y, Zhou Y, Yang L, Huang H and Hu Y: Off-the-Shelf, multiplexed-engineered iPSC-derived CD33 CAR-NK cells for treatment of acute myeloid leukemia. Blood. 140:126852022. View Article : Google Scholar

106 

Shapiro RM and Romee R: iPSC-derived CD19 CAR NK cells for relapsed or refractory lymphoma. Lancet. 405:98–99. 2025. View Article : Google Scholar : PubMed/NCBI

107 

Ghobadi A, Bachanova V, Patel K, Park JH, Flinn I, Riedell PA, Bachier C, Diefenbach CS, Wong C, Bickers C, et al: Induced pluripotent stem-cell-derived CD19-directed chimeric antigen receptor natural killer cells in B-cell lymphoma: A phase 1, first-in-human trial. Lancet. 405:127–136. 2025. View Article : Google Scholar : PubMed/NCBI

108 

Merino A, Maakaron J and Bachanova V: Advances in NK cell therapy for hematologic malignancies: NK source, persistence and tumor targeting. Blood Rev. 60:1010732023. View Article : Google Scholar : PubMed/NCBI

109 

Bachanova V, Cayci Z, Lewis D, Maakaron JE, Janakiram M, Bartz A, Payne S, Wong C, Cooley S, Valamehr B, et al: Initial clinical activity of FT596, a first-in-class, multi-antigen targeted, off-the-shelf, iPSC-Derived CD19 CAR NK cell therapy in Relapsed/Refractory B-Cell lymphoma. Blood. 136:82020. View Article : Google Scholar

110 

Bachanova V, Deol A, Al-Juhaishi TMS, Lulla PD, Byrne MT, Wong C, Bickers C, Greene T, Wong L, Villa B, et al: Safety and efficacy of FT522, a first-in-class, multi-antigen targeted, off-the-shelf, iPSC-Derived CD19 CAR NK cell therapy with alloimmune defense receptor (ADR) in Relapsed/Refractory B-Cell lymphoma. Blood. 144:65432024. View Article : Google Scholar

111 

Patel K, Namburi S, Latif T and Oluwole OO: Interim results from the ELiPSE-1 study: A phase 1, multicenter, open-label study of CNTY-101 in subjects with relapsed or refractory CD19-positive B-cell malignancies. J Clin Oncol. 42:70232024. View Article : Google Scholar

112 

Borges L, Wallet MA, Bullaughey CL, Naso MF, Gurung B, Keating S, Carton JM, Wheeler JC, Campion L, Mendonca M, et al: Development of multi-engineered iPSC-derived CAR-NK cells for the treatment of B-cell malignancies. Blood. 138:17292021. View Article : Google Scholar

113 

Sermer D, Elavalakanar P, Abramson JS, Palomba ML, Salles G and Arnason J: Targeting CD19 for diffuse large B cell lymphoma in the era of CARs: Other modes of transportation. Blood Rev. 57:1010022023. View Article : Google Scholar : PubMed/NCBI

114 

Ramachandran I, Rothman S, Clausi M, McFadden K, Salantes B, Jih G, Brigman T, Kelly S, Hall MS, Yee S, et al: Multiple doses of Cnty-101, an iPSC-derived allogeneic CD19 targeting CAR-NK product, are safe and result in tumor microenvironment changes associated with response: A case study. Blood. 142:16542023. View Article : Google Scholar

115 

Terren I, Orrantia A, Vitalle J, Zenarruzabeitia O and Borrego F: NK cell metabolism and tumor microenvironment. Front Immunol. 10:22782019. View Article : Google Scholar : PubMed/NCBI

116 

Xu Z, Yang J, Xin X, Liu C, Li L, Mei X and Li M: Merits and challenges of iPSC-derived organoids for clinical applications. Front Cell Dev Biol. 11:11889052023. View Article : Google Scholar : PubMed/NCBI

117 

Huyghe M, Desterke C, Imeri J, Belliard N, Chaker D, Oudrirhi N, Bezerra H, Turhan AG, Bennaceur-Griscelli A and Griscelli F: Comparative analysis of iPSC-derived NK cells from two differentiation strategies reveals distinct signatures and cytotoxic activities. Front Immunol. 15:14637362024. View Article : Google Scholar : PubMed/NCBI

118 

Nianias A and Themeli M: Induced pluripotent stem cell (iPSC)-derived lymphocytes for adoptive cell immunotherapy: recent advances and challenges. Curr Hematol Malig Rep. 14:261–268. 2019. View Article : Google Scholar : PubMed/NCBI

119 

Zhou Y, Cheng L, Liu L and Li X: NK cells are never alone: Crosstalk and communication in tumour microenvironments. Mol Cancer. 22:342023. View Article : Google Scholar : PubMed/NCBI

120 

Linke JA, Munn LL and Jain RK: Compressive stresses in cancer: Characterization and implications for tumour progression and treatment. Nat Rev Cancer. 24:768–791. 2024. View Article : Google Scholar : PubMed/NCBI

121 

Xie D, Zhu S and Bai L: Lactic acid in tumor microenvironments causes dysfunction of NKT cells by interfering with mTOR signaling. Sci China Life Sci. 59:1290–1296. 2016. View Article : Google Scholar : PubMed/NCBI

122 

Gao Y, Zhou H, Liu G, Wu J, Yuan Y and Shang A: Tumor microenvironment: lactic acid promotes tumor development. J Immunol Res. 2022:31193752022. View Article : Google Scholar : PubMed/NCBI

123 

Miao L, Lu C, Zhang B, Li H, Zhao X, Chen H, Liu Y and Cui X: Advances in metabolic reprogramming of NK cells in the tumor microenvironment on the impact of NK therapy. J Transl Med. 22:2292024. View Article : Google Scholar : PubMed/NCBI

124 

Lozada JR, Zhang B, Miller JS and Cichocki F: NK cells from human cytomegalovirus-seropositive individuals have a distinct metabolic profile that correlates with elevated mTOR signaling. J Immunol. 211:539–550. 2023. View Article : Google Scholar : PubMed/NCBI

125 

Osuna-Espinoza KY and Rosas-Taraco AG: Metabolism of NK cells during viral infections. Front Immunol. 14:10641012023. View Article : Google Scholar : PubMed/NCBI

126 

Yin Y, Feng W, Chen J, Chen X, Wang G, Wang S, Xu X, Nie Y, Fan D, Wu K and Xia L: Immunosuppressive tumor microenvironment in the progression, metastasis, and therapy of hepatocellular carcinoma: From bench to bedside. Exp Hematol Oncol. 13:722024. View Article : Google Scholar : PubMed/NCBI

127 

Wang Q, Shao X, Zhang Y, Zhu M, Wang FXC, Mu J, Li J, Yao H and Chen K: Role of tumor microenvironment in cancer progression and therapeutic strategy. Cancer Med. 12:11149–11165. 2023. View Article : Google Scholar : PubMed/NCBI

128 

Arner EN and Rathmell JC: Metabolic programming and immune suppression in the tumor microenvironment. Cancer Cell. 41:421–433. 2023. View Article : Google Scholar : PubMed/NCBI

129 

Tong L, Jimenez-Cortegana C, Tay AHM, Wickstrom S, Galluzzi L and Lundqvist A: NK cells and solid tumors: Therapeutic potential and persisting obstacles. Mol Cancer. 21:2062022. View Article : Google Scholar : PubMed/NCBI

130 

Vivier E, Rebuffet L, Narni-Mancinelli E, Cornen S, Igarashi RY and Fantin VR: Natural killer cell therapies. Nature. 626:727–736. 2024. View Article : Google Scholar : PubMed/NCBI

131 

Tarannum M, Romee R and Shapiro RM: Innovative strategies to improve the clinical application of NK cell-based immunotherapy. Front Immunol. 13:8591772022. View Article : Google Scholar : PubMed/NCBI

132 

Carreira-Santos S, Lopez-Sejas N, Gonzalez-Sanchez M, Sánchez-Hernández E, Pera A, Hassouneh F, Durán E, Solana R, Casado JG and Tarazona R: Enhanced expression of natural cytotoxicity receptors on cytokine-induced memory-like natural killer cells correlates with effector function. Front Immunol. 14:12564042023. View Article : Google Scholar : PubMed/NCBI

133 

Wang Z, Guan D, Wang S, Chai LYA, Xu S and Lam KP: Glycolysis and oxidative phosphorylation play critical roles in natural killer cell receptor-mediated natural killer cell functions. Front Immunol. 11:2022020. View Article : Google Scholar : PubMed/NCBI

134 

Poznanski SM, Barra NG, Ashkar AA and Schertzer JD: Immunometabolism of T cells and NK cells: Metabolic control of effector and regulatory function. Inflamm Res. 67:813–828. 2018. View Article : Google Scholar : PubMed/NCBI

135 

Shankar K, Zingler-Hoslet I, Tabima DM, Zima S, Shi L, Gimse K, Forsberg MH, Katta V, Davis SZ, Maldonado D, et al: Virus-free CRISPR knockin of a chimeric antigen receptor into KLRC1 generates potent GD2-specific natural killer cells. Mol Ther. 33:1014–1030. 2024. View Article : Google Scholar

136 

Hsu J, Hodgins JJ, Marathe M, Nicolai CJ, Bourgeois-Daigneault MC, Trevino TN, Azimi CS, Scheer AK, Randolph HE, Thompson TW, et al: Contribution of NK cells to immunotherapy mediated by PD-1/PD-L1 blockade. J Clin Invest. 128:4654–4668. 2018. View Article : Google Scholar : PubMed/NCBI

137 

Iwai Y, Ishida M, Tanaka Y, Okazaki T, Honjo T and Minato N: Involvement of PD-L1 on tumor cells in the escape from host immune system and tumor immunotherapy by PD-L1 blockade. Proc Natl Acad Sci USA. 99:12293–12297. 2002. View Article : Google Scholar : PubMed/NCBI

138 

Clerico M, Ragaini S and Cavallo F: Non-Hodgkin lymphoma treated with anti-CD20 antibody-based immunochemotherapy. Resistance to Anti-Cd20 Antibodies and Approaches for their Reversal Elsevier. 103–122. 2024. View Article : Google Scholar

139 

Hoffman B and Liebermann DA: Apoptotic signaling by c-MYC. Oncogene. 27:6462–6472. 2008. View Article : Google Scholar : PubMed/NCBI

140 

Ding S: Therapeutic reprogramming toward regenerative medicine. Chem Rev. 125:1805–1822. 2025. View Article : Google Scholar : PubMed/NCBI

141 

Agostini A, Orlacchio A, Carbone C and Guerriero I: Understanding tricky cellular and molecular interactions in pancreatic tumor microenvironment: New food for thought. Front Immunol. 13:8762912022. View Article : Google Scholar : PubMed/NCBI

Related Articles

Journal Cover

August-2025
Volume 32 Issue 2

Print ISSN: 1791-2997
Online ISSN:1791-3004

Sign up for eToc alerts

Recommend to Library

Copy and paste a formatted citation
x
Spandidos Publications style
Wei X, Su C, Liu Y, Wei N, Xiang K, Qian Q and Xu Z: IPSC‑derived NK cells for immunotherapy and therapeutic perspective (Review). Mol Med Rep 32: 222, 2025.
APA
Wei, X., Su, C., Liu, Y., Wei, N., Xiang, K., Qian, Q., & Xu, Z. (2025). IPSC‑derived NK cells for immunotherapy and therapeutic perspective (Review). Molecular Medicine Reports, 32, 222. https://doi.org/10.3892/mmr.2025.13587
MLA
Wei, X., Su, C., Liu, Y., Wei, N., Xiang, K., Qian, Q., Xu, Z."IPSC‑derived NK cells for immunotherapy and therapeutic perspective (Review)". Molecular Medicine Reports 32.2 (2025): 222.
Chicago
Wei, X., Su, C., Liu, Y., Wei, N., Xiang, K., Qian, Q., Xu, Z."IPSC‑derived NK cells for immunotherapy and therapeutic perspective (Review)". Molecular Medicine Reports 32, no. 2 (2025): 222. https://doi.org/10.3892/mmr.2025.13587