Next Article in Journal
Unraveling the Heterogeneity of ALS—A Call to Redefine Patient Stratification for Better Outcomes in Clinical Trials
Previous Article in Journal
A Possible Therapeutic Application of the Selective Inhibitor of Urate Transporter 1, Dotinurad, for Metabolic Syndrome, Chronic Kidney Disease, and Cardiovascular Disease
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Building a Better Defense: Expanding and Improving Natural Killer Cells for Adoptive Cell Therapy

by
Andreia Maia
1,2,3,
Mubin Tarannum
2,
Joana R. Lérias
4,
Sara Piccinelli
2,
Luis Miguel Borrego
5,6,
Markus Maeurer
4,7,
Rizwan Romee
2 and
Mireia Castillo-Martin
1,8,*
1
Molecular and Experimental Pathology Laboratory, Champalimaud Centre for the Unknown, Champalimaud Foundation, 1400-038 Lisbon, Portugal
2
NK Cell Gene Manipulation and Therapy Laboratory, Division of Cellular Therapy and Stem Cell Transplant, Dana–Farber Cancer Institute, Harvard Medical School, Boston, MA 02115, USA
3
NOVA Medical School, NOVA University of Lisbon, 1099-085 Lisbon, Portugal
4
ImmunoTherapy/ImmunoSurgery, Champalimaud Centre for the Unknown, Champalimaud Foundation, 1400-038 Lisbon, Portugal
5
Comprehensive Health Research Centre (CHRC), NOVA Medical School, Faculdade de Ciências Médicas (FCM), NOVA University of Lisbon, 1099-085 Lisbon, Portugal
6
Immunoallergy Department, Hospital da Luz, 1600-209 Lisbon, Portugal
7
I Medical Clinic, University of Mainz, 55131 Mainz, Germany
8
Pathology Service, Champalimaud Clinical Center, Champalimaud Foundation, 1400-038 Lisbon, Portugal
*
Author to whom correspondence should be addressed.
Cells 2024, 13(5), 451; https://doi.org/10.3390/cells13050451
Submission received: 11 February 2024 / Revised: 1 March 2024 / Accepted: 1 March 2024 / Published: 5 March 2024
(This article belongs to the Special Issue Advances in the Study of Natural Killer (NK) Cells)

Abstract

:
Natural killer (NK) cells have gained attention as a promising adoptive cell therapy platform for their potential to improve cancer treatments. NK cells offer distinct advantages over T-cells, including major histocompatibility complex class I (MHC-I)-independent tumor recognition and low risk of toxicity, even in an allogeneic setting. Despite this tremendous potential, challenges persist, such as limited in vivo persistence, reduced tumor infiltration, and low absolute NK cell numbers. This review outlines several strategies aiming to overcome these challenges. The developed strategies include optimizing NK cell expansion methods and improving NK cell antitumor responses by cytokine stimulation and genetic manipulations. Using K562 cells expressing membrane IL-15 or IL-21 with or without additional activating ligands like 4-1BBL allows “massive” NK cell expansion and makes multiple cell dosing and “off-the-shelf” efforts feasible. Further improvements in NK cell function can be reached by inducing memory-like NK cells, developing chimeric antigen receptor (CAR)-NK cells, or isolating NK-cell-based tumor-infiltrating lymphocytes (TILs). Memory-like NK cells demonstrate higher in vivo persistence and cytotoxicity, with early clinical trials demonstrating safety and promising efficacy. Recent trials using CAR-NK cells have also demonstrated a lack of any major toxicity, including cytokine release syndrome, and, yet, promising clinical activity. Recent data support that the presence of TIL-NK cells is associated with improved overall patient survival in different types of solid tumors such as head and neck, colorectal, breast, and gastric carcinomas, among the most significant. In conclusion, this review presents insights into the diverse strategies available for NK cell expansion, including the roles played by various cytokines, feeder cells, and culture material in influencing the activation phenotype, telomere length, and cytotoxic potential of expanded NK cells. Notably, genetically modified K562 cells have demonstrated significant efficacy in promoting NK cell expansion. Furthermore, culturing NK cells with IL-2 and IL-15 has been shown to improve expansion rates, while the presence of IL-12 and IL-21 has been linked to enhanced cytotoxic function. Overall, this review provides an overview of NK cell expansion methodologies, highlighting the current landscape of clinical trials and the key advancements to enhance NK-cell-based adoptive cell therapy.

Graphical Abstract

1. What Are Natural Killer Cells?

Natural killer (NK) cells are large granular lymphocytes representing 5–10% of circulating lymphocytes in healthy adults, playing a crucial role in recognizing and eliminating transformed and infected cells [1]. Distinguished by the absence of CD3 and CD19 and the presence of CD56, NKp46, and NKp80 expression, NK cells consist of two principal subsets: the immature or regulatory CD56bright CD16+/− cells and the mature or cytotoxic CD56dim CD16+ cells (Figure 1A) [1,2,3]. The CD56bright subset, more prevalent in the lymph nodes, secretes cytokines and chemokines in an inflammatory milieu, recruiting and modulating immune cells such as neutrophils, macrophages, T-cells, B-cells, and dendritic cells (Figure 1B) [4,5,6]. However, CD56bright cells can be rapidly primed to acquire potent cytotoxic function upon cytokine activation [7]. In contrast, the CD56dim subset exhibits higher direct cell cytotoxicity against tumor targets, mediating antibody-dependent cell cytotoxicity (ADCC) through the CD16 expression, which binds to the Fc region of IgG1 antibodies (Figure 1C) [8,9,10]. Furthermore, both NK cell subsets induce tumor cell apoptosis by expressing death ligands like FasL and TRAIL (Figure 1C) [11].

2. How Do NK Cells Modulate Their Immune Response?

NK cells exhibit a broad array of activating and inhibitory receptors, influencing their function [12]. Activating receptors include C-type lectin receptors, like CD94/NKG2C (binding to HLA-E) and CD94/NKG2D (binding to MHC-I chain-related molecules A/B (MIC-A/B)); natural cytotoxic receptors (NCR), including NKp30 (binding to hemagglutinin (HA), B7-H6, galectin-3, and glycosaminoglycans (GAGs)), NKp44 (binding to HA, GAGs, and NKp44L), and NKp46 (binding to HA, GAGs, and ecto-calreticulin); Fc receptor (FcγR, mediating ADCC); DNAM-1 (binding to poliovirus receptor (PVR) and Nectin-2); and activating killer-cell immunoglobulin-like receptors (KIR), such as KIR-2DS and KIR-3DS (recognizing MHC-I molecules) [12,13,14,15,16,17,18,19,20,21]. Inhibitory receptors include inhibitory KIR, such as KIR-2DL and KIR-3DL (recognizing MHC-I molecules), and C-type lectin receptors like CD94/NKG2A/B (binding to HLA-E) [12,13].
NK cell activation is mediated by a balance between activating and inhibitory signals (Figure 2). The “missing-self” hypothesis proposes that loss or downregulation of MHC-I molecules on tumor cells leads to decreased inhibitory signals, thus favoring NK cell activation [22]. In contrast, “stress-induced” activation occurs when stress ligands such as MIC-A/B are upregulated on infected or malignant cells, engaging activating receptors like NKG2D on NK cells [12,23]. In addition, NK cell function can be restrained by immune checkpoints, including NKG2A, TIM-3, TIGIT, and CD96 [24]. Although not highly prevalent, programmed death-1 (PD-1) expression has been observed in NK cells, thereby inducing dysfunction of the NK cells within the tumors with increased PD-L1 expression [25,26].

3. What Are the Major Advantages of NK Cells and Their Respective Sources for Adoptive Cell Therapy?

Chimeric antigen receptor (CAR)-T cells have significantly advanced cellular immunotherapy for cancer treatment [27,28,29,30]. Other T-cell-based approaches, including tumor-infiltrating lymphocytes (TIL) and T-cell receptor (TCR) T-cells, have also demonstrated promising efficacy, leading to the approval of several products by the FDA [30,31,32,33,34,35]. Nevertheless, T-cell-based therapies are associated with a relatively high risk of developing cytokine release syndrome (CRS), immune effector cell-associated neurotoxicity syndrome (ICANS), and hemophagocytic lymphohistiocytosis (HLH), resulting in prolonged cytopenia and risk of graph-versus-host disease (GVHD) in an allogeneic setting [36,37,38,39]. In contrast, NK/CAR-NK-cell-based products are not associated with these side effects, making them an attractive alternative [40]. In addition, the versatility of NK cell sources, including peripheral blood (PB), cord blood (CB), hematopoietic stem cells (HSC), and induced pluripotent stem cells (iPSC), increases their accessibility, supporting NK-cell-based products as a viable alternative to T-cells [41,42]. The major advantages and disadvantages of NK cell sources are described below (Figure 3).

3.1. Peripheral Blood (PB)

NK cells constitute approximately 5 to 10% of total lymphocytes in PB, exhibiting a mature phenotype with high cytotoxicity against tumors (Figure 3A) [1]. Within the PB-NK cells, the CD56bright subset accounts for approximately 10%, while the remainder is mostly CD56dim NK cells [43]. NK cells are commonly isolated from peripheral blood mononuclear cells (PBMC), obtained through leukapheresis, followed by a bead-based selection process [44]. Despite being readily available, using PB as a source for NK cells has several limitations, including relatively low cell numbers and donor-dependent variability [44].

3.2. Cord Blood (CB)

NK cells constitute around 23% of CB cells, with similar proportions of the CD56bright and CD56dim subsets found in PB (Figure 3B) [45]. CB-NK cells exhibit lower levels of adhesion molecules (CD2, CD11a, CD18, and CD54) and maturation receptors (KIR and CD57) while maintaining similar expression of the key cytotoxic molecules like granzyme B and perforin compared to PB-NK cells [46,47,48]. Although CB presents a limited number of NK cells per CB unit, recent efficient expansion strategies (described in the following section) have allowed the generation of several infusion products [49,50,51]. Alternatively, NK cells can be differentiated from the CD34+ HSC that are highly enriched in the CB and display many similarities to PB-NK cells, though with a lower inhibitory receptor expression [52,53,54,55].

3.3. Embryonic and Induced Pluripotent Stem Cells

Recent advances have allowed efficient differentiation of human embryonic stem cells (hESCs) and induced pluripotent stem cells (iPSCs) into highly functional NK cells (iNK cells), sharing many functional and phenotypic similarities (Figure 3C) [56,57,58,59]. iPSC-NK cells express CD56, DNAM-1, CD69, NKG2A/D, and NCR, like PB-NK cells [60,61,62]. However, the CD16 expression is lower, impacting their ability to mediate ADCC, while their KIR expression is variable, with certain cell populations expressing high levels of KIR and others not expressing [60,61,62,63]. Additionally, iNK cells generated from hESC expressed CD16 and KIR and lysed malignant cells by direct cell-mediated cytotoxicity and ADCC [55,62,64]. However, while iPSC-NK cells present a potential advantage for “off-the-shelf” manufacturing, their scale-up is somewhat more challenging than hESC-NK cells [59,65,66].
Figure 3. Sources of NK cells. (A) In PB-NK cells represent 5–10% of total lymphocytes, with approximately 90% being CD56dim CD16+ NK cells and the remaining 10% comprising CD56bright CD16+/− NK cells. (B) CB-NK cells can be obtained from CB or by differentiating CD34+ cells through exposure to specific cytokines. NK cells comprise 23% of the total cells in CB, and the proportions of CB-NK cell subsets are similar to PBMC-NK cells. (C) Human embryonic stem cells (hESCs) and induced pluripotent stem cells (iPSCs) can generate NK cells with functional and phenotype similarities to PB-NK cells, although presenting a lower CD16 expression.
Figure 3. Sources of NK cells. (A) In PB-NK cells represent 5–10% of total lymphocytes, with approximately 90% being CD56dim CD16+ NK cells and the remaining 10% comprising CD56bright CD16+/− NK cells. (B) CB-NK cells can be obtained from CB or by differentiating CD34+ cells through exposure to specific cytokines. NK cells comprise 23% of the total cells in CB, and the proportions of CB-NK cell subsets are similar to PBMC-NK cells. (C) Human embryonic stem cells (hESCs) and induced pluripotent stem cells (iPSCs) can generate NK cells with functional and phenotype similarities to PB-NK cells, although presenting a lower CD16 expression.
Cells 13 00451 g003

4. What Are the Major NK Cell Expansion Approaches?

Despite the availability of multiple sources for generating clinical-grade NK cell products, their low absolute numbers remain a limitation [67,68]. Several expansion methods have been developed to address this, with the major ones being summarized below [57,69,70,71,72,73,74,75,76,77,78,79,80,81,82,83,84,85,86,87,88].

4.1. Cytokine-Based Expansion

Cytokines such as IL-2, IL-12, IL-15, IL-18, and IL-21 play a crucial role in activating and regulating NK cell biology and, therefore, have been commonly used in various expansion methods [69,70]. IL-2 stimulates the PI3K/AKT pathway, promoting cell survival and proliferation via mTOR activation [89]. Sharing similar downstream pathways, IL-15 induces cell survival, activation, cytolytic activity, and cytokine production [90]. In contrast to IL-15, IL-2 also activates T-cells, including regulatory T-cells (Tregs) [91,92]. IL-12 enhances cytokine production, such as IFN-γ, and promotes cytotoxicity, while IL-18 sustains cell survival and exhibits co-stimulatory effects with other activation signals [93,94]. IL-21 supports NK cell survival and enhances cytotoxicity by upregulating granzymes and perforin molecules [95]. Various cytokine-based expansion protocols are highlighted in Table 1.
PBMC cultured with IL-2 achieved a 193-fold expansion of NK cells within 21 days [71]. IL-2, OK432, and an anti-CD16 monoclonal antibody (mAb) yielded a high NK cell purity of 76.9% with expansion folds ranging from hundreds to thousands, albeit with T-cell expansion [72]. Isolated NK cells cultured with IL-2 initially showed decreased NK cell counts by day 4, but recovered afterwards [73]. IL-15 demonstrated potential for NK cell expansion, while IL-21 triggered maturation and functionality, especially with short-term stimulation [74]. When expanded with IL-2, IL-15, anti-CD3 monoclonal antibody, tacrolimus, and dalteparin sodium, CB-NK cells achieved over 1700-fold expansion with more than 70% purity [75].

4.2. Feeder-Cell-Based Expansion

Feeder-cell-based expansion systems, typically involving immortalized or tumor cell lines in combination with cytokines, have become a common approach for expanding and activating NK/CAR-NK cells (Table 2) [76,77,78,96]. Using Epstein–Barr Virus-Immortalized Lymphoblastoid cell line (EBV-LCL) as feeder cells resulted in a 53-fold expansion of NK cells after one week with IL-21 stimulation and superior NK cell cytotoxicity [76]. In addition, CB-NK cells cultured with EBV-LCL cells showed an up to 6092-fold expansion after 35 days [77]. Furthermore, NK cells from patients with advanced cancer were cultured with NK-cell-depleted-PBMC from diseased and healthy donors [87]. A higher NK cell expansion rate was observed when using healthy donors’ feeder cells (300-fold) compared to diseased feeder cells (164.9-fold) [87].
Genetic alterations of feeder cells have allowed even higher NK cell expansion rates [57,79,80,81,82,83,84,85,97,98]. For instance, K562 cells expressing MICA, 4-1BBL, and soluble IL-15 induced a 550-fold cell expansion rate, increasing the NK cell percentage from 14.8% to 86.7% in PBMC [79]. NK cells cultured with K562 incorporating membrane-bound IL-15 (mbIL15) and 4-1BBL (K562-mbIL15-41BBL) induced a 277-fold expansion [80]. CB-NK cells cultured with K562-mbIL15-41BBL cells reached a 35-fold cell expansion and demonstrated superior cytotoxicity [81]. However, NK cells cultured with K562-mbIL15 cells displayed short telomeres [79]. In contrast, K562 cells overexpressing mbIL-21 (K562-mbIL21) promoted a massive log-phase NK cell expansion without evidence of senescence for up to 6 weeks [82]. Moreover, iNK cells cultured and expanded with K562-mbIL21-41BBL cells displayed 95.8% purity (CD3CD56+ phenotype) [57]. A human B-lymphoblastoid cell line, 721.221, expressing mbIL21, induced superior NK cell expansion, purity, and cytotoxicity compared to NK cells cultured with K562-mbIL21 cells [83]. Additionally, K562-HLA-E feeder cells induced more than 10,000-fold expansion of NKG2C+ memory-like adaptive NK cells [97,98]. Furthermore, K562 cells expressing OX40 ligand (K562-OX40L) with IL-21 stimulation induced approximately 2000-fold NK cell expansion compared to 303-fold when using only K562 cells [84].
Feeder cells from autologous PBMC have also been successfully used to expand NK cells [78,86,87,88]. NK cells reached 62.7-fold expansion when cultured with irradiated autologous NK cell-depleted PMBC as feeder cells for 19 days [86]. In addition, CB-derived CD56+ cytotoxic cells cultured with IL-2 and irradiated autologous lymphocytes reached 156.3-fold expansion on day 26 [99]. Using autologous CD56-depleted feeder cells induced a 212-fold expansion of NK cells compared to 22.5-fold when using only IL-2 [88]. Additionally, culturing PBMC with irradiated autologous stimulated T-cells (FN-CH296) induced 90% purity and an NK cell median expansion of 4720-fold [78].
Table 2. Analysis of NK cell expansion rates across diverse feeder-cell-based expansion methods, varying in source, media, feeder cells, duration, and culture material.
Table 2. Analysis of NK cell expansion rates across diverse feeder-cell-based expansion methods, varying in source, media, feeder cells, duration, and culture material.
Ref.Cell SourceCulture FactorsFeedersTime (Days)Culture MaterialResults
[76]PBMC-NK cellsTexMACS containing 5% HS type AB, 500 U/mL of IL-2, 100 ng/mL of IL-21, and feeder cells.EBV-LCL cells (ratio 1:20).7N/ANK cells reached 22-fold expansion in 1 week, which increased to 53-fold with IL-21.
[77]CB-NK cellsX-VIVO 20 media with 10% heat-inactivated human AB serum, 500 IU/mL rhIL-2 and 2 mM GlutaMAX-1.EBV-LCL cells. Up to 40T75 flasks.CD3 CD56+ cells reached a median of 6092-fold expansion.
[87]Isolated NK cellsAIM-V medium supplemented with 5% HS, 1000 U/mL of IL-2 and OKT3.NK cell—negative fraction of PBMC.1424-well plates.The patient’s NK cells co-cultured with healthy donor feeder cells reached 300-fold expansion.
[79]PBMC or purified NK cellsMedia with 10 ng/mL IL-2.Genetically modified K562 cells.2424-well plates.K562-MICA-41BBL-IL-15 cells induced 550-fold NK cell expansion in 24 days.
[80]PBMC or purified NK cellsRPMI-1640 media with 10 IU/mL human IL-2 and 10% FBS.K562-mb15-41BBL cells.2124-well plates.PBMC showed a 21.6-fold expansion of NK cells, while a 277-fold was reached in purified NK cells after 21 days.
[81]CB-NK cellsRPMI-1640 media with 10% FBS and 10 IU/mL of recombinant IL-2.K562-mbIL15-41BBL cells.1424-well plates.K562-mbIL15-41BBL cells induced a 35-fold NK cell expansion.
[82]PBMCRPMI-1640 media with 50 IU/mL IL-2, 10% FBS, L-glutamine, and P/S.K562-mbIL21 cells.42T75 flasks.K562-mbIL21 cells induced a 47,967-fold expansion of NK cells by day 21.
[57]iNK cellsB0 media supplemented with cytokines.K562-IL21-4-1BBL cells.42 At day 42, 98.5% of cells had a CD3 CD56+ phenotype and reached 105 to 106-fold expansion.
[83]PBMCRPMI-1640 media with 10% FBS, 2mM L-glutamine, 100 U/mL P/S, 100 U/mL IL-2 and 5 ng/mL of IL-15.221-mbIL21 cells.20G-Rex 6 Multiwell cell culture plates.NK cells showed a 39,663-fold increase with 221-mbIL21 cells compared to a 3588-fold expansion with K562-mbIL21 cells.
[84]PBMCRPMI 1640 media with 10% FBS, P/S, 4 mmol/L of L-glutamine, and 10 U/mL of IL-2. After one week, IL-2 was increased to 100 U/mL, and 5 ng/mL of IL-15 was added. IL-21 was added at different concentrations.K562-OX40L cells.3824-well plateAfter four weeks, K562-OX40L cells and short exposure to IL-21 induced a 2000-fold expansion of NK cells.
[85]Purified NK cellsAIM-V media with cytokines or feeder cells and 100 ng/mL OKT3 in the first culture cycle.NK92-Neo2/15-OX40L cells.21N/ANK92-Neo2/15-OX40L cells induced a 2180-fold increase of NK cells after 21 days without additional cytokines.
[86]Isolated NK cellsCellGro SCGM with 5% HS, P/S and 10 ng/mL of OKT3. 200 U/mL of IL-2 was added alone or with 10 ng/mL of IL-15.Autologous feeder cells.19Baxter LifeCell culture bags.NK cells reached 62.7-fold expansion with IL-2 and 117-fold when IL-15 was added.
[99]CB-NK cellsRPMI-1640 media with IL-2, 10% human AB, 1 mM L-glutamina, 10 U/mL Pen, and 0.01 mg/mL of streptomycin.Autologous PBMC.2624-well plate.CB-CD56+ cytotoxic NK cells reached 156.3-fold increased on day 26.
[88]Isolated NK cellsTexMACS media with 5% human AB serum, 1000 U/mL of IL-2, and 10 ng/mL of OKT3.Autologous CD56-depleted PBMC.1224-well plate, T25 and T75 flasks.NK cells reached a 212-fold expansion with feeder cells, while only IL-2 showed a 22.5-fold expansion.
[78]PBMCGT-T507 with 1% plasma, IL-2, and OK-432.FN-CH296 cells.21–22Flasks and culture bags.A median of 4720-fold expansion was reached after 22 days with 90.96% purity.
Antibody (Ab), cord blood (CB), fetal bovine serum (FBS), human serum (HS), interleukin (IL), international unit (IU), natural killer (NK) cells, penicillin/streptomycin (P/S), peripheral blood mononuclear cell (PBMC), stem cell growth medium (SCGM), unit (U).

4.3. Culture Materials Used for NK Cell Expansion

Selecting appropriate culture materials, including flasks, bags, or bioreactors, is crucial in ensuring efficient NK cell expansion. Effector cells, particularly CD4+, CD8+, CD8+ CD56+ T-cells, and CD56+ NK cells cultured in bags and flasks, exhibited no significant differences in expansion, phenotype, or function over a 7-day period [100]. In contrast, NK cells reached a 530-fold expansion in bags compared to 1100-fold using flasks [101]. Culture in flasks carries the risk of exposure and contamination, which can be minimized in a GMP laboratory [101]. On the other hand, culturing in bags requires maintaining a certain cell concentration, and the gas exchange is restricted by the media’s volume, limiting the supply of nutrients and impacting NK cell proliferation [102]. Bioreactors are considered the most practical method despite being more expensive due to minimal hands-on time. However, bioreactors typically require higher starting cell numbers, and the expansion rate might be lower than other methods [101].

5. How Do Cell Culture Strategies Improve NK Cell Activity?

Although naturally cytotoxic, NK cells’ activity can be further improved during cell culture. The previous section described several expansion methods, while the current section outlines the changes in phenotype, persistence, and cytotoxicity that NK cells undergo during expansion.

5.1. Phenotype

NK cells display diverse activating and inhibitory receptors, and their expression often changes during cell culture [103]. Stimulation with IL-2, OK432, and anti-CD16 antibody upregulated CD16 and NKG2D, while IL-2 and IL-21 increased TRAIL, NKG2D, and DNAM-1 expression [72,76,78]. Combining IL-2 and IL-15 increased NKG2D and NKp44 levels, with no significant changes observed for NKp30, NKp46, and DNAM-1 [86]. NK cells cultured with PBMC (as feeder cells) showed a high frequency of RANKL, B7-H3, and HLA class II, particularly HLA-DR [88]. K562-MICA-41BBL-IL15 cells increased the expression of CD69, CD16, NKG2D, and CXCR3 on NK cells [79]. Despite higher NK cell expansion rates with K562-mbIL21 cells versus K562-mbIL15, NK cells exhibited a similar phenotype and cytotoxicity, maintaining donor KIR repertoire and showing high NCR, CD16, and NKG2D expression [82].

5.2. Telomere Length

Telomere shortening is a limiting factor for NK cell expansion [104]. Exposure to K562-mb15-41BBL cells limited proliferation due to telomere shortening [104]. To overcome this, NK cells overexpressing human telomerase reverse transcriptase (TERT) demonstrated an extended lifespan, maintaining a high percentage of cells in the S/G2 phase [105]. TERT-NK cells cultured with K562-mb15-41BBL cells showed prolonged proliferation for over a year, maintaining a normal karyotype and genotype [106]. NK cells in culture with K562 cells and IL-2 stimulation showed higher phosphorylation of STAT3, an activator of human TERT [73,107]. In addition, NK cells expanded with K562-mbIL21 cells, demonstrated increased telomere length, upregulation of STAT3, and reduced senescence [82].

5.3. Cytotoxicity

Stimulation with cytokines and feeder cells improves NK cell antitumor responses [108]. IL-2 and IL-12 promoted IFN-γ production, while IL-15 and brief IL-21 exposure boosted NK cell cytotoxicity, degranulation, and cytokine secretion [74,109]. Although K562-41BBL-IL15 cells promoted NK cell cytotoxicity and IFN-γ production, using K562-MICA-41BBL-IL15 induced a stronger effect [79]. Additionally, K562-mbIL21-41BBL cells also improved IFN-γ and TNF-α production [82]. NK-92-OX40L cells secreting neoleukin-2/15 (Neo-2/15) increased NK cell cytotoxicity against tumor cells [84].
Expansion strategies influence NK cell phenotype, function, and cytotoxicity, determining the efficacy of NK-cell-based therapies.

6. Which Other Strategies Improve NK Cell Antitumor Response?

Despite improvements in NK cell expansion and cytotoxicity during cell culture, their clinical application is hampered by major challenges, including short half-life, limited tumor infiltration, and low in vivo persistence [110]. Key strategies to further enhance NK-cell-based immunotherapeutic approaches include stimulation of memory-like NK cells, genetic manipulation including CAR-NK cells, and, more recently, TIL-NK cells [111,112,113].

6.1. Memory-like NK Cells

NK cells can acquire memory-like properties in response to cytokine stimulation, particularly IL-12, IL-15, and IL-18, generating what is called the cytokine-induced memory-like (CIML) NK cells or, in the context of viral infections like cytomegalovirus (CMV), inducing what is called the adaptive NK cells [112]. CIML NK cells undergo transcriptional, epigenetic, and metabolic reprogramming, leading to increased proliferation, cytotoxicity, and long-term persistence in mouse models and patients [114,115,116]. Upregulation of phosphorylated STAT5 and demethylated conserved noncoding sequence 1 (CNS1) was found in memory-like NK cells [114,117]. In addition, increased expressions of IL-2Rα (CD25), nutrient transporters, including transferrin receptor (CD71), amino acid transporter (CD98), and glucose transporters (GLUT1 and GLUT3) were observed [118,119,120]. These cells demonstrated enhanced function, proliferation, and in vivo persistence against cancer cells [112,121,122]. On the other hand, under CMV reactivation, adaptive NK cells demonstrate inferior NKG2A, NKp30, and NKp46 expression while increasing NKG2C, KIR, and CD57 markers [123,124]. Adaptive NK cells are characterized by a memory-like phenotype and increased cytotoxicity, producing more IFN-γ molecules with long-term persistence, making them attractive for immunotherapy [115,123].

6.2. Chimeric Antigen Receptor (CAR) Technology

CAR therapy involves modifying immune cells to express a synthetic receptor binding to a tumor-associated antigen like CD19, expressed by B-cells (both normal and B-cell lymphomas), and BCMA, expressed by normal and malignant plasma cells [29,125,126]. While T-cells are the most widely used immune cell for CAR therapy, they present several limitations, such as the alloreactivity and GVHD risks, making autologous T-cells a preferential choice [38,127]. In contrast, CAR-NK cells, derived from allogeneic sources, provide an “off-the-shelf” product without inducing GVHD, avoiding the massive release of cytokines and neurotoxicity [51,128].
CD19-CAR-CIML NK cells demonstrated enhanced IFN-γ production, degranulation, and specific killing against NK-resistant lymphoma cell lines [129]. Recently, CD19-CAR-CB NK cells expressing IL-15 demonstrated safety and efficacy in CD19+ B-cell malignancies [40]. In this study, 73% of patients had a response to the treatment without toxicities observed, including CRS, GVHD, and neurotoxicity [40,51]. CAR-CIML NK cells targeting a neoepitope generated in nucleophosmin-1 (NPM1)-mutated acute myeloid lymphoma (AML) displayed potent activity, improving AML outcomes in xenograft models [130]. Various strategies have recently been explored to improve CAR-NK cells, including CISH knockout, promoting metabolic fitness, cell expansion, antitumor cytotoxicity, IL-15-mediated STAT signaling, and superior NCR expression [131,132,133]. In addition, enhanced NK cell functions against glioblastoma through αv integrin blockade, TGF-β inhibition, and CRISPR gene editing of the TGFBR2 gene were also reported [134]. CD38 knockout NK cells overcome the daratumumab-induced fratricide, improving AML and multiple myeloma (MM) treatment efficacy [135]. Overexpression of CXCR1 in NKG2D-CAR-NK cells enhanced their migration toward in vitro and in vivo tumors [136]. Similarly, overexpression of CXCR4 or CCR7 in NK cells improved migration and infiltration into specific tissues, reducing tumor burden and extending survival in mice [137,138].

6.3. Tumor-Infiltrating Lymphocytes (TILs), including TIL-NK Cells

TILs primarily comprise T-cells that have migrated into a tumor [139]. TIL therapy has shown significant clinical results, particularly in melanoma patients [140,141,142]. Furthermore, ongoing studies in breast and colorectal cancers highlight the potential of TILs as a source of antigen-specific immune cells [113,143,144]. Recent studies have emphasized that TILs include NK cells (TIL-NK cells), associated with improved prognosis in multiple malignancies [145,146,147,148,149,150]. In lung cancer, TIL-NK cells are predominantly CD56bright perforinlow, exhibiting lower cytotoxicity but with similar cytokine production compared to the PB-NK cells [151]. In soft tissue sarcoma, NK cells represent around 20% of the TIL population [152]. In comparison, less than 0.5% of TIL-NK cells are found in pancreatic ductal adenocarcinoma (PDAC), attributed to the low expression of the chemokine receptor CXCR2 [153]. Transgenic expression of CXCR2 facilitates NK cell infiltration, although proliferation is limited in the hypoxic TME [154]. CCR7 expression, involved in lymphocyte migration to lymph nodes, remains unchanged, while CXCR3, mediating NK cell recruitment to tumor sites, is enhanced in expanded NK cells [79]. Additionally, CXCL9 and CXCL10 expression in the TME and IL-15 stimulation promote the recruitment of NK cells and cytotoxic CD8+ T-cells via CXCR3 into the tumors [155].
While until now, no method has been explicitly published for the expansion of TIL-NK cells, it is crucial to consider the promising prospects of TIL-NK-cell-based therapies in cancer [149,156]. Despite the limited cell numbers of TIL-NK cells, their superior tumor-infiltration capacities and association with improved overall survival of cancer patients underscore significant potential [149,156]. Additionally, the recent FDA approval of Amtagvi, an autologous TIL therapy for advanced melanoma in adults, emphasizes the promising potential of TIL therapy as a treatment option for cancer [157,158].

7. Which Are Currently the Major NK-Cell-Based Clinical Trials?

The current clinical application of NK cells includes autologous and allogeneic NK-cell-based approaches [159]. So far, the NK cell clinical trials have predominantly focused on patients with hematological diseases, though promising data from recent preclinical studies strongly support their evaluation in solid tumors [160,161,162]. The following section summarizes key clinical trials, highlighting the advantages of expanded NK cells.

7.1. Autologous NK Cells

In the autologous setting, NK cells have been safely infused and expanded in vivo with IL-2 administration; however, their efficacy has been limited [78,163,164]. In a phase I trial, autologous NK cells expanded with K562-mbIL15-41BBL cells reported stable disease combined with trastuzumab in 6 of 19 patients with HER2-positive malignancies [163]. Infusion of activated autologous NK cells into MM patients post-autologous HCT also supported the broader feasibility of this therapy [165]. Despite the prolonged survival of ex vivo IL-2-activated autologous NK cells in preclinical studies, no clinical responses were observed in patients with metastatic melanoma or renal cell carcinoma [166]. Their minimal clinical activity can be due to a lack of KIR/ligand mismatch in the autologous tumor cells and/or due to their limited in vivo persistence after adoptive transfer [166]. These challenges represent a significant hurdle for autologous NK-cell-based therapy [166]. Several ongoing clinical trials are evaluating autologous NK cell therapy for hepatocellular carcinoma (NCT06044506) and MM in combination with low IL-2 (NCT04634435).

7.2. Allogeneic NK Cells

The use of allogeneic NK cells has been associated with inducing remission and preventing relapse in AML and MM patients [167]. Using KIR-mismatched donor NK cells after haploidentical HCT demonstrated a significantly reduced risk of relapse in high-risk AML [168,169]. Subsequently, allogeneic NK cells from unrelated healthy donors were assessed in advanced lymphoma and solid tumors [160]. Among 17 patients, 47.1% showed stable disease, highlighting the safety and the potential efficacy of administrating random-donor allogeneic NK cells and thus expanding cell donor options [160]. In another study, the use of haploidentical PBMC (CD3+ T-cell-depleted and NK-cell-enriched) was safe and induced complete response (CR) in 5 of 19 poor-prognosis AML patients [170]. Similarly, IL-2-activated allogeneic NK cells combined with anti-CD20 mAb yielded responses in 14 of 15 relapsed/refractory CD20+ lymphoma patients in a phase II clinical trial [171].
While haploidentical NK cell infusion induced remissions, the presence of Tregs may have contributed to their diminished efficacy [172]. Depletion of host Tregs with IL-2-diphtheria fusion protein improved the efficacy of haploidentical NK cell therapy, resulting in higher donor NK cell expansion in relapsed-refractory AML patients [172]. Substituting IL-2 with IL-15 showed promising results, with 36% of patients exhibiting robust in vivo NK cell expansion and 32% achieving CR, avoiding Treg stimulation [173]. In a phase II trial, IL-15 administered subcutaneously (SC) resulted in NK cell expansion in 27% of the patients, and 40% achieved remission [173]. However, while IL-15 improved in vivo NK cell expansion and remission rates, it was also associated with previously unreported CRS after SC administration [173]. Moreover, IL-15 superagonist complex ALT-803 was well tolerated, stimulating NK and CD8+ T-cells without increasing Tregs [174].
Considering the high risk of relapse after allogeneic HCT, donor-derived CIML NK cells are attractive in myeloid malignancies that have relapsed after haploidentical HCT [175]. A first-in-human phase I trial with donor CIML NK cells in relapsed or refractory AML reported four of nine patients achieving CR and one achieving morphologic leukemia-free state (MLFS), resulting in an overall response rate of 55% and a CR rate of 45% [122]. Additionally, CIML NK cell infusion led to rapid 10- to 50-fold expansion in vivo, sustained over months, supporting CIML NK cells as a platform for post-transplant relapse myeloid disease treatment [175]. These findings highlight the importance of expanding and stimulating NK cells before infusion, whether as CIML or conventional NK cells, to improve immunotherapy efficacy.

7.3. Allogeneic CB-NK Cells

In a first-in-human trial, CB-NK cells, expanded with IL-2 and K562-mbIL21 cells, were infused in MM patients receiving high-dose chemotherapy and autologous HCT [176]. This trial demonstrated safety and efficacy, with 10 patients achieving at least a good partial response, including 8 near CR [176]. Additionally, in recurrent ovarian carcinoma, CB-NK cells exhibited safety and in vivo expansion capacity [177]. The “off-shelf” CB-NK cell product, oNKord, has obtained approval for AML patients [178]. The phase I trial demonstrated safety and efficacy in elderly AML patients, ineligible for allogeneic HCT, while an ongoing phase II trial is evaluating oNKord in patients with minimal residue disease (MRD) (NCT04632316) [178]. Recently, a phase I/II trial of CB-NK cells expressing CD19-CAR and IL-15 was evaluated in patients with CD19+ B-cell malignancies [40,51]. The 1-year overall survival (OR) and progression-free survival were 68% and 32%, respectively, with patients achieving OR correlated with higher levels and longer persistence of CAR-NK cells [40]. No notable toxicities were observed, including CRS and GvHD [40].

8. What Is the Future of NK-Cell-Based Therapy?

While NK cells hold great promise, challenges remain, including low in vivo persistence and proliferation capacity, which can compromise their effectiveness in cancer therapy [179]. This review aims to elucidate several strategies used to improve NK cell proliferation and antitumor function, either through expansion methodologies, cytokine stimulation, or genetic modifications. Diverse culture methods are explored using various cytokines (such as IL-2, IL-12, IL-15, IL-18, and IL-21) and feeder cells (including genetically modified or nongenetically modified cell lines, as well as autologous or allogeneic cells), with emphasis on their effects on NK cell expansion, phenotype, telomere length, and cytotoxic activity. Notably, stimulation with IL-2 and IL-15 has been shown to promote NK cell expansion, while IL-12 and IL-21 have been associated with enhancing cell cytotoxicity [71,72,73,74]. In addition, using genetically engineered feeder cells, like K562-mbIL21 cells, has demonstrated remarkable ex vivo NK cell expansion capacities [82,180].
Furthermore, the generation of memory-like NK cells, characterized by superior IFN-γ production and cytotoxicity, represents another strategy to improve in vivo persistence, providing a sustained and potent functional alternative [122]. On the other hand, genetic manipulation of NK cells to develop CAR-NK cells has shown promising results, enhancing NK cell targeting without inducing severe side effects [40,51,128,129,136,137]. Moreover, exploring TIL therapy, particularly TIL-NK cells, presents a compelling alternative to improve tumor treatment, given their association with improved overall survival in cancer patients [149,156]. Recently, FDA approval of an autologous TIL therapy for advanced melanoma has underscored the promising antitumor potential of TILs [157,158].
Despite the promising outcomes in cancer, especially in hematological malignancies, translating NK-cell-based therapies in solid tumors faces significant challenges, with poor tumor trafficking and highly immunosuppressive TME as major barriers for both NK- and non-NK-cell-based cellular therapy approaches [110]. The TME can induce NK cell dysfunction and exhaustion through various mechanisms, including suppressive immune or nonimmune cells (e.g., Tregs and myeloid-derived suppressor cells (MDSC)), cytokines like TGF-β, overexpression of inhibitory ligands (e.g., HLA-E), and downregulation of activating ligands [181]. Several strategies are being evaluated to overcome these challenges, including exploring TIL’s superior tumor infiltration capacity, and developing novel CAR-engineered cells [182,183,184]. Engineered NK cells with synthetic receptors, sustained cytokine production, safety mechanisms like drug-inducible suicide genes, or “on/off switches” through small molecule administrations promise great potential [185]. Targeting cancer and other cells in the TME that help tumor growth, like cancer-associated fibroblasts (CAFs), can potentially improve NK-cell-based approaches.
Furthermore, the most effective NK-cell-based immunotherapy may involve combining them with other immune cells, such as CAR-T cells, TIL, or CAR-macrophages, taking advantage of the unique strengths of each approach to enhance tumor control significantly. Additionally, combining NK cell therapies with immune checkpoint inhibitors might be a great strategy, especially considering the dysfunction and exhaustion markers expressed by NK and T-cells within the TME, such as CD161, TIGIT, and CD96 [152]. Lastly, there is a huge interest in developing the in vivo “arming” of NK and other immune cells to mitigate labor-intensive and expensive adoptive cell therapies [186]. Recent progress in RNA-based approaches combined with nanoparticle-based technology makes in vivo modulation of the immune cells feasible. However, challenges like low transfection efficacy, short RNA half-life, and limited cell specificity remain the major barriers to these efforts [187].
In summary, this review offers insights into strategies aimed at enhancing NK cell function through expansion methodologies or genetic modifications, elucidating their impact on NK cell phenotype, proliferation, and cytotoxicity. In addition, this review highlights the current clinical trial landscape and key advancements aimed at further enhancing NK-cell-based adoptive cell therapy.

Author Contributions

A.M. and M.C.-M. drafted the main text, figures, and tables. A.M. drew the figures. M.C.-M., J.R.L., L.M.B., M.M., M.T., S.P. and R.R. reviewed and revised the text, providing comments, and additional scientific information. All authors have read and agreed to the published version of the manuscript.

Funding

This work was funded by the Fundação da Ciência e Tecnologia (FCT) with reference number SFRH/BD/144965/2019 and by the European Hematology Association (EHA) Research Mobility Grant 2023.

Conflicts of Interest

The authors declare no potential conflicts of interest.

References

  1. Del Zotto, G.; Antonini, F.; Pesce, S.; Moretta, F.; Moretta, L.; Marcenaro, E. Comprehensive Phenotyping of Human PB NK Cells by Flow Cytometry. J. Quant. Cell Sci. 2020, 97, 891–899. [Google Scholar] [CrossRef] [PubMed]
  2. Lam, R.A.; Chwee, J.Y.; Le Bert, N.; Sauer, M.; Pogge Von Strandmann, E.; Gasser, S. Regulation of Self-Ligands for Activating Natural Killer Cell Receptors. Ann. Med. 2013, 45, 384–394. [Google Scholar] [CrossRef] [PubMed]
  3. Cui, F.; Qu, D.; Sun, R.; Nan, K. Circulating CD16+CD56+ Nature Killer Cells Indicate the Prognosis of Colorectal Cancer after Initial Chemotherapy. Med. Oncol. 2018, 36, 84. [Google Scholar] [CrossRef] [PubMed]
  4. Ferlazzo, G.; Münz, C. NK Cell Compartments and Their Activation by Dendritic Cells. J. Immunol. 2004, 172, 1333–1339. [Google Scholar] [CrossRef] [PubMed]
  5. Wang, F.; Lau, J.K.C.; Yu, J. The Role of Natural Killer Cell in Gastrointestinal Cancer: Killer or Helper. Oncogene 2021, 40, 717–730. [Google Scholar] [CrossRef]
  6. Barnes, S.A.; Trew, I.; De Jong, E.; Foley, B. Making a Killer: Selecting the Optimal Natural Killer Cells for Improved Immunotherapies. Front. Immunol. 2021, 12, 765705. [Google Scholar] [CrossRef] [PubMed]
  7. Wagner, J.A.; Rosario, M.; Romee, R.; Berrien-Elliott, M.M.; Schneider, S.E.; Leong, J.W.; Sullivan, R.P.; Jewell, B.A.; Becker-Hapak, M.; Schappe, T.; et al. CD56bright NK Cells Exhibit Potent Antitumor Responses Following IL-15 Priming. J. Clin. Investig. 2017, 127, 4042–4058. [Google Scholar] [CrossRef] [PubMed]
  8. Bald, T.; Krummel, M.F.; Smyth, M.J.; Barry, K.C. The NK Cell–Cancer Cycle: Advances and New Challenges in NK Cell–Based Immunotherapies. Nat. Immunol. 2020, 21, 835–847. [Google Scholar] [CrossRef]
  9. Valipour, B.; Velaei, K.; Abedelahi, A.; Karimipour, M.; Darabi, M.; Charoudeh, H.N. NK Cells: An Attractive Candidate for Cancer Therapy. J. Cell. Physiol. 2019, 234, 19352–19365. [Google Scholar] [CrossRef]
  10. Lo Nigro, C.; Macagno, M.; Sangiolo, D.; Bertolaccini, L.; Aglietta, M.; Carlo Merlano, M. NK-Mediated Antibody-Dependent Cell-Mediated Cytotoxicity in Solid Tumors: Biological Evidence and Clinical Perspectives. Ann. Transl. Med. 2019, 7, 105. [Google Scholar] [CrossRef]
  11. Judge, S.J.; Murphy, W.J.; Canter, R.J. Characterizing the Dysfunctional NK Cell: Assessing the Clinical Relevance of Exhaustion, Anergy, and Senescence. Front. Cell. Infect. Microbiol. 2020, 10, 49. [Google Scholar] [CrossRef]
  12. Fang, F.; Xiao, W.; Tian, Z. NK Cell-Based Immunotherapy for Cancer. Semin. Immunol. 2017, 31, 37–54. [Google Scholar] [CrossRef] [PubMed]
  13. Meckawy, G.R.; Mohamed, A.M.; Zaki, W.K.; Khattab, M.A.; Amin, M.M.; ElDeeb, M.A.; El-Najjar, M.R.; Safwat, N.A. Natural Killer NKG2A and NKG2D in Patients with Colorectal Cancer. J. Gastrointest. Oncol. 2019, 10, 218–225. [Google Scholar] [CrossRef] [PubMed]
  14. Chester, C.; Fritsch, K.; Kohrt, H.E. Natural Killer Cell Immunomodulation: Targeting Activating, Inhibitory, and Co-Stimulatory Receptor Signaling for Cancer Immunotherapy. Front. Immunol. 2015, 6, 601. [Google Scholar] [CrossRef] [PubMed]
  15. Béziat, V.; Hilton, H.G.; Norman, P.J.; Traherne, J.A. Deciphering the Killer-Cell Immunoglobulin-like Receptor System at Super-Resolution for Natural Killer and T-Cell Biology. Immunology 2017, 150, 248–264. [Google Scholar] [CrossRef] [PubMed]
  16. Zingoni, A.; Ardolino, M.; Santoni, A.; Cerboni, C. NKG2D and DNAM-1 Activating Receptors and Their Ligands in NK-T Cell Interactions: Role in the NK Cell-Mediated Negative Regulation of T Cell Responses. Front. Immunol. 2012, 3, 408. [Google Scholar] [CrossRef] [PubMed]
  17. Dhar, P.; Wu, J.D. NKG2D and Its Ligands in Cancer. Curr. Opin. Immunol. 2018, 51, 55–61. [Google Scholar] [CrossRef] [PubMed]
  18. Gumá, M.; Busch, L.K.; Salazar-Fontana, L.I.; Bellosillo, B.; Morte, C.; García, P.; López-Botet, M. The CD94/NKG2C Killer Lectin-like Receptor Constitutes an Alternative Activation Pathway for a Subset of CD8+ T Cells. Eur. J. Immunol. 2005, 35, 2071–2080. [Google Scholar] [CrossRef] [PubMed]
  19. Barrow, A.D.; Martin, C.J.; Colonna, M. The Natural Cytotoxicity Receptors in Health and Disease. Front. Immunol. 2019, 10, 909. [Google Scholar] [CrossRef]
  20. Littera, R.; Piredda, G.; Argiolas, D.; Lai, S.; Congeddu, E.; Ragatzu, P.; Melis, M.; Carta, E.; Michittu, M.B.; Valentini, D.; et al. KIR and Their HLA Class I Ligands: Two More Pieces towards Completing the Puzzle of Chronic Rejection and Graft Loss in Kidney Transplantation. PLoS ONE 2017, 12, e0180831. [Google Scholar] [CrossRef]
  21. Sen Santara, S.; Lee, D.-J.; Crespo, Â.; Hu, J.J.; Walker, C.; Ma, X.; Zhang, Y.; Chowdhury, S.; Meza-Sosa, K.F.; Lewandrowski, M.; et al. The NK Cell Receptor NKp46 Recognizes Ecto-Calreticulin on ER-Stressed Cells. Nature 2023, 616, 348–356. [Google Scholar] [CrossRef] [PubMed]
  22. Ljunggren, H.; Kirre, K. The Search of the ‘Missing Self’: MHC Molecules NK Cell Recognition. Immunol. Today 1990, 11, 237–244. [Google Scholar] [CrossRef]
  23. Long, E.O.; Rajagopalan, S. Stress Signals Activate Natural Killer Cells. J. Exp. Med. 2002, 196, 1399–1402. [Google Scholar] [CrossRef]
  24. Khan, M.; Arooj, S.; Wang, H.; Wang, H. NK Cell-Based Immune Checkpoint Inhibition. Front. Genet. 2020, 11, 167. [Google Scholar] [CrossRef] [PubMed]
  25. Hsu, J.; Hodgins, J.J.; Marathe, M.; Nicolai, C.J.; Bourgeois-Daigneault, M.C.; Trevino, T.N.; Azimi, C.S.; Scheer, A.K.; Randolph, H.E.; Thompson, T.W.; et al. Contribution of NK Cells to Immunotherapy Mediated by PD-1/PD-L1 Blockade. J. Clin. Investig. 2018, 128, 4654–4668. [Google Scholar] [CrossRef]
  26. Concha-Benavente, F.; Kansy, B.; Moskovitz, J.; Moy, J.; Chandran, U.; Ferris, R.L. PD-L1 Mediates Dysfunction in Activated PD-1 þ NK Cells in Head and Neck Cancer Patients. Cancer Immunol. Res. 2018, 6, 1548–1560. [Google Scholar] [CrossRef]
  27. Neelapu, S.S.; Locke, F.L.; Bartlett, N.L.; Lekakis, L.J.; Miklos, D.B.; Jacobson, C.A.; Braunschweig, I.; Oluwole, O.O.; Siddiqi, T.; Lin, Y.; et al. Axicabtagene Ciloleucel CAR T-Cell Therapy in Refractory Large B-Cell Lymphoma. N. Engl. J. Med. 2017, 377, 2531–2544. [Google Scholar] [CrossRef]
  28. Schuster, S.J.; Svoboda, J.; Chong, E.A.; Nasta, S.D.; Mato, A.R.; Anak, Ö.; Brogdon, J.L.; Pruteanu-Malinici, I.; Bhoj, V.; Landsburg, D.; et al. Chimeric Antigen Receptor T Cells in Refractory B-Cell Lymphomas. N. Engl. J. Med. 2017, 377, 2545–2554. [Google Scholar] [CrossRef] [PubMed]
  29. Sterner, R.C.; Sterner, R.M. CAR-T Cell Therapy: Current Limitations and Potential Strategies. Blood Cancer J. 2021, 11, 69. [Google Scholar] [CrossRef]
  30. Maude, S.L.; Laetsch, T.W.; Buechner, J.; Rives, S.; Boyer, M.; Bittencourt, H.; Bader, P.; Verneris, M.R.; Stefanski, H.E.; Myers, G.D.; et al. Tisagenlecleucel in Children and Young Adults with B-Cell Lymphoblastic Leukemia. N. Engl. J. Med. 2018, 378, 439–448. [Google Scholar] [CrossRef]
  31. Locke, F.L.; Ghobadi, A.; Jacobson, C.A.; Miklos, D.B.; Lekakis, L.J.; Oluwole, O.O.; Lin, Y.; Braunschweig, I.; Hill, B.T.; Timmerman, J.M.; et al. Long-Term Safety and Activity of Axicabtagene Ciloleucel in Refractory Large B-Cell Lymphoma (ZUMA-1): A Single-Arm, Multicentre, Phase 1–2 Trial. Lancet Oncol. 2019, 20, 31–42. [Google Scholar] [CrossRef]
  32. Abramson, J.S.; Palomba, M.L.; Gordon, L.I.; Lunning, M.A.; Wang, M.; Arnason, J.; Mehta, A.; Purev, E.; Maloney, D.G.; Andreadis, C.; et al. Lisocabtagene Maraleucel for Patients with Relapsed or Refractory Large B-Cell Lymphomas (TRANSCEND NHL 001): A Multicentre Seamless Design Study. Lancet 2020, 396, 839–852. [Google Scholar] [CrossRef] [PubMed]
  33. Wang, M.; Munoz, J.; Goy, A.; Locke, F.L.; Jacobson, C.A.; Hill, B.T.; Timmerman, J.M.; Holmes, H.; Jaglowski, S.; Flinn, I.W.; et al. KTE-X19 CAR T-Cell Therapy in Relapsed or Refractory Mantle-Cell Lymphoma. N. Engl. J. Med. 2020, 382, 1331–1342. [Google Scholar] [CrossRef] [PubMed]
  34. Munshi, N.C.; Anderson, L.D.; Shah, N.; Madduri, D.; Berdeja, J.; Lonial, S.; Raje, N.; Lin, Y.; Siegel, D.; Oriol, A.; et al. Idecabtagene Vicleucel in Relapsed and Refractory Multiple Myeloma. N. Engl. J. Med. 2021, 384, 705–716. [Google Scholar] [CrossRef] [PubMed]
  35. Martin, T.; Usmani, S.Z.; Berdeja, J.G.; Agha, M.; Cohen, A.D.; Hari, P.; Avigan, D.; Deol, A.; Htut, M.; Lesokhin, A.; et al. Ciltacabtagene Autoleucel, an Anti-B-Cell Maturation Antigen Chimeric Antigen Receptor T-Cell Therapy, for Relapsed/Refractory Multiple Myeloma: CARTITUDE-1 2-Year Follow-Up. J. Clin. Oncol. 2022, 41, 1265–1274. [Google Scholar] [CrossRef] [PubMed]
  36. Watanabe, N.; Mo, F.; McKenna, M.K. Impact of Manufacturing Procedures on CAR T Cell Functionality. Front. Immunol. 2022, 13, 876339. [Google Scholar] [CrossRef] [PubMed]
  37. Brudno, J.N.; Kochenderfer, J.N. Toxicities of Chimeric Antigen Receptor T Cells: Recognition and Management. Blood 2016, 127, 3321–3330. [Google Scholar] [CrossRef] [PubMed]
  38. Hirayama, A.V.; Turtle, C.J. Toxicities of CD19 CAR-T Cell Immunotherapy. Am. J. Hematol. 2019, 94, 42–49. [Google Scholar] [CrossRef] [PubMed]
  39. Allen, E.S.; Stroncek, D.F.; Ren, J.; Eder, A.F.; West, K.A.; Fry, T.J.; Lee, D.W.; Mackall, C.L.; Conry-Cantilena, C. Autologous Lymphapheresis for the Production of Chimeric Antigen Receptor T Cells. Transfusion 2017, 57, 1133–1141. [Google Scholar] [CrossRef]
  40. Marin, D.; Li, Y.; Basar, R.; Rafei, H.; Daher, M.; Dou, J.; Mohanty, V.; Dede, M.; Nieto, Y.; Uprety, N.; et al. Safety, Efficacy and Determinants of Response of Allogeneic CD19-Specific CAR-NK Cells in CD19+ B Cell Tumors: A Phase 1/2 Trial. Nat. Med. 2024. [Google Scholar] [CrossRef]
  41. Aptsiauri, N.; Ruiz-Cabello, F.; Garrido, F. The Transition from HLA-I Positive to HLA-I Negative Primary Tumors: The Road to Escape from T-Cell Responses. Curr. Opin. Immunol. 2018, 51, 123–132. [Google Scholar] [CrossRef]
  42. Garrido, F.; Aptsiauri, N.; Doorduijn, E.M.; Garcia Lora, A.M.; van Hall, T. The Urgent Need to Recover MHC Class I in Cancers for Effective Immunotherapy. Curr. Opin. Immunol. 2016, 39, 44–51. [Google Scholar] [CrossRef] [PubMed]
  43. Angelo, L.S.; Banerjee, P.P.; Monaco-Shawver, L.; Rosen, J.B.; Makedonas, G.; Forbes, L.R.; Mace, E.M.; Orange, J.S. Practical NK Cell Phenotyping and Variability in Healthy Adults. Immunol. Res. 2015, 62, 341–356. [Google Scholar] [CrossRef]
  44. Laskowski, T.J.; Biederstädt, A.; Rezvani, K. Natural Killer Cells in Antitumour Adoptive Cell Immunotherapy. Nat. Rev. Cancer 2022, 22, 557–575. [Google Scholar] [CrossRef]
  45. Sarvaria, A.; Jawdat, D.; Madrigal, J.A.; Saudemont, A. Umbilical Cord Blood Natural Killer Cells, Their Characteristics, and Potential Clinical Applications. Front. Immunol. 2017, 8, 329. [Google Scholar] [CrossRef]
  46. Dalle, J.; Menezes, J.; Wagner, É.; Blagdon, M.; Champagne, J.; Champagne, M.A.; Duval, M.; Hematology-oncology, D.P. Characterization of Cord Blood Natural Killer Cells: Implications for Transplantation and Neonatal Infections. Pediatr. Res. 2005, 57, 649–655. [Google Scholar] [CrossRef] [PubMed]
  47. Tanaka, H.; Kai, S.; Yamaguchi, M.; Misawa, M.; Fujimori, Y.; Yamamoto, M.; Hara, H. Analysis of Natural Killer ( NK ) Cell Activity and Adhesion Molecules on NK Cells from Umbilical Cord Blood. Eur. J. Haematol. 2003, 71, 29–38. [Google Scholar] [CrossRef]
  48. Fan, Y.; Yang, B.; Wu, C. Phenotypic and Functional Heterogeneity of Natural Killer Cells from Umbilical Cord Blood Mononuclear Cells. Immunol. Investig. 2008, 37, 79–96. [Google Scholar] [CrossRef] [PubMed]
  49. Horwitz, M.E.; Stiff, P.J.; Cutler, C.; Brunstein, C.; Hanna, R.; Mariarz, R.T.; Rezvani, A.R.; Karris, N.A.; McGuirk, J.; Valcarcel, D.; et al. Omidubicel vs Standard Myeloablative Umbilical Cord Blood Transplantation: Results of a Phase 3 Randomized Study. Blood 2021, 138, 1429–1440. [Google Scholar] [CrossRef] [PubMed]
  50. Shpall, E.J.; Rezvani, K. Cord Blood Expansion Has Arrived. Blood 2021, 138, 1381–1382. [Google Scholar] [CrossRef]
  51. Liu, E.; Marin, D.; Banerjee, P.; MacApinlac, H.A.; Thompson, P.; Basar, R.; Kerbauy, L.N.; Overman, B.; Thall, P.; Kaplan, M.; et al. Use of CAR-Transduced Natural Killer Cells in CD19-Positive Lymphoid Tumors. N. Engl. J. Med. 2020, 382, 545–553. [Google Scholar] [CrossRef] [PubMed]
  52. Spanholtz, J.; Preijers, F.; Tordoir, M.; Trilsbeek, C.; Paardekooper, J.; de Witte, T.; Schaap, N.; Dolstra, H. Clinical-Grade Generation of Active NK Cells from Cord Blood Hematopoietic Progenitor Cells for Immunotherapy Using a Closed-System Culture Process. PLoS ONE 2011, 6, e20740. [Google Scholar] [CrossRef] [PubMed]
  53. Cany, J.; van der Waart, A.B.; Spanholtz, J.; Tordoir, M.; Jansen, J.H.; van der Voort, R.; Schaap, N.M.; Dolstra, H. Combined IL-15 and IL-12 Drives the Generation of CD34+-Derived Natural Killer Cells with Superior Maturation and Alloreactivity Potential Following Adoptive Transfer. Oncoimmunology 2015, 4, e1017701. [Google Scholar] [CrossRef] [PubMed]
  54. Herrera, L.; Salcedo, J.M.; Santos, S.; Vesga, M.Á.; Borrego, F.; Eguizabal, C. OP9 Feeder Cells Are Superior to M2-10B4 Cells for the Generation of Mature and Functional Natural Killer Cells from Umbilical Cord Hematopoietic Progenitors. Front. Immunol. 2017, 8, 755. [Google Scholar] [CrossRef] [PubMed]
  55. Luevano, M.; Madrigal, A.; Saudemont, A. Generation of Natural Killer Cells from Hematopoietic Stem Cells in Vitro for Immunotherapy. Cell. Mol. Immunol. 2012, 9, 310–320. [Google Scholar] [CrossRef] [PubMed]
  56. Li, Y.; Hermanson, D.L.; Moriarity, B.S.; Kaufman, D.S.; Li, Y.; Hermanson, D.L.; Moriarity, B.S.; Kaufman, D.S. Human IPSC-Derived Natural Killer Cells Engineered with Chimeric Antigen Receptors Enhance Anti-Tumor Activity. Cell Stem Cell 2018, 23, 181–192.e5. [Google Scholar] [CrossRef] [PubMed]
  57. Cichocki, F.; Bjordahl, R.; Gaidarova, S.; Mahmood, S.; Abujarour, R.; Wang, H.; Tuininga, K.; Felices, M.; Davis, Z.B.; 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. 2020, 12, eaaz5618. [Google Scholar] [CrossRef]
  58. Knorr, D.A.; Bock, A.; Brentjens, R.J.; Kaufman, D.S. Engineered Human Embryonic Stem Cell-Derived Lymphocytes to Study in Vivo Trafficking and Immunotherapy. Stem Cells Dev. 2013, 22, 1861–1869. [Google Scholar] [CrossRef]
  59. Knorr, D.A.; Ni, Z.; Hermanson, D.L.; Hexum, M.K.; Bendzick, L.; Cooper, L.J.; Lee, D.A.; Kaufman, D.S. Clinical-Scale Derivation of Natural Killer Cells From Human Pluripotent Stem Cells for Cancer Therapy. Stem Cells Transl. Med. 2013, 2, 274–283. [Google Scholar] [CrossRef]
  60. Goldenson, B.H.; Hor, P.; Kaufman, D.S. IPSC-Derived Natural Killer Cell Therapies—Expansion and Targeting. Front. Immunol. 2022, 13, 841107. [Google Scholar] [CrossRef]
  61. Euchner, J.; Sprissler, J.; Cathomen, T.; Fürst, D.; Schrezenmeier, H.; Debatin, K.M.; Schwarz, K.; Felgentreff, K. Natural Killer Cells Generated From Human Induced Pluripotent Stem Cells Mature to CD56brightCD16+NKp80+/− In-Vitro and Express KIR2DL2/DL3 and KIR3DL1. Front. Immunol. 2021, 12, 640672. [Google Scholar] [CrossRef] [PubMed]
  62. Woll, P.S.; Martin, C.H.; Miller, J.S.; Kaufman, D.S. Human Embryonic Stem Cell-Derived NK Cells Acquire Functional Receptors and Cytolytic Activity. J. Immunol. 2005, 175, 5095–5103. [Google Scholar] [CrossRef] [PubMed]
  63. Goldenson, B.H.; Zhu, H.; Wang, Y.Z.M.; Heragu, N.; Bernareggi, D.; Ruiz-Cisneros, A.; Bahena, A.; Ask, E.H.; Hoel, H.J.; Malmberg, K.J.; et al. Umbilical Cord Blood and IPSC-Derived Natural Killer Cells Demonstrate Key Differences in Cytotoxic Activity and KIR Profiles. Front. Immunol. 2020, 11, 561553. [Google Scholar] [CrossRef]
  64. Woll, P.S.; Grzywacz, B.; Tian, X.; Marcus, R.K.; Knorr, D.A.; Verneris, M.R.; Kaufman, D.S. Human Embryonic Stem Cells Differentiate into a Homogeneous Population of Natural Killer Cells with Potent In Vivo Antitumor Activity. Blood 2009, 113, 6094–6101. [Google Scholar] [CrossRef]
  65. Eguizabal, C.; Zenarruzabeitia, O.; Monge, J.; Santos, S.; Vesga, M.A.; Maruri, N.; Arrieta, A.; Riñón, M.; Tamayo-orbegozo, E.; Amo, L.; et al. Natural Killer Cells for Cancer Immunotherapy: Pluripotent Stem Cells-Derived NK Cells as an Immunotherapeutic Perspective. Front. Immunol. 2014, 5, 439. [Google Scholar] [CrossRef]
  66. Maddineni, S.; Silberstein, J.L.; Sunwoo, J.B. Emerging NK Cell Therapies for Cancer and the Promise of next Generation Engineering of IPSC-Derived NK Cells. J. Immunother. Cancer 2022, 10, e004693. [Google Scholar] [CrossRef] [PubMed]
  67. Nayar, S.; Dasgupta, P.; Galustian, C. Extending the Lifespan and Efficacies of Immune Cells Used in Adoptive Transfer for Cancer Immunotherapies—A Review. Oncoimmunology 2015, 4, e1002720. [Google Scholar] [CrossRef]
  68. Zhang, Y.; Wallace, D.L.; De Lara, C.M.; Ghattas, H.; Asquith, B.; Worth, A.; Griffin, G.E.; Taylor, G.P.; Tough, D.F.; Beverley, P.C.L.; et al. In Vivo Kinetics of Human Natural Killer Cells: The Effects of Ageing and Acute and Chronic Viral Infection. Immunology 2007, 121, 258–265. [Google Scholar] [CrossRef]
  69. Leong, J.W.; Chase, J.M.; Romee, R.; Schneider, S.E.; Sullivan, R.P.; Cooper, M.A.; Fehniger, T.A. Preactivation with IL-12, IL-15, and IL-18 Induces Cd25 and a Functional High-Affinity Il-2 Receptor on Human Cytokine-Induced Memory-like Natural Killer Cells. Biol. Blood Marrow Transplant. 2014, 20, 463–473. [Google Scholar] [CrossRef]
  70. Heinze, A.; Grebe, B.; Bremm, M.; Huenecke, S.; Munir, T.A.; Graafen, L.; Frueh, J.T.; Merker, M.; Rettinger, E.; Soerensen, J.; et al. The Synergistic Use of IL-15 and IL-21 for the Generation of NK Cells From CD3/CD19-Depleted Grafts Improves Their Ex Vivo Expansion and Cytotoxic Potential Against Neuroblastoma: Perspective for Optimized Immunotherapy Post Haploidentical Stem Cell Transplantation. Front. Immunol. 2019, 10, 2816. [Google Scholar] [CrossRef]
  71. Carlens, S.; Gilljam, M.; Chambers, B.J.; Aschan, J.; Guven, H.; Ljunggren, H.; Christensson, B.; Dilber, M.S. A New Method for In Vitro Expansion of Cytotoxic Human CD3−CD56+ Natural Killer Cells. Hum. Immunol. 2001, 62, 1092–1098. [Google Scholar] [CrossRef]
  72. Deng, X.; Terunuma, H.; Nieda, M.; Xiao, W.; Nicol, A. Synergistic Cytotoxicity of Ex Vivo Expanded Natural Killer Cells in Combination with Monoclonal Antibody Drugs against Cancer Cells. Int. Immunopharmacol. 2012, 14, 593–605. [Google Scholar] [CrossRef]
  73. Koehl, U.; Brehm, C.; Huenecke, S.; Zimmermann, S.Y.; Kloess, S.; Bremm, M.; Ullrich, E.; Soerensen, J.; Quaiser, A.; Erben, S.; et al. Clinical Grade Purification and Expansion of NK Cell Products for an Optimized Manufacturing Protocol. Front. Oncol. 2013, 3, 118. [Google Scholar] [CrossRef]
  74. Wagner, J.; Pfannenstiel, V.; Waldmann, A.; Bergs, J.W.J.; Brill, B.; Huenecke, S.; Klingebiel, T.; Rödel, F.; Buchholz, C.J.; Wels, W.S.; et al. A Two-Phase Expansion Protocol Combining Interleukin (IL)-15 and IL-21 Improves Natural Killer Cell Proliferation and Cytotoxicity against Rhabdomyosarcoma. Front. Immunol. 2017, 8, 676. [Google Scholar] [CrossRef] [PubMed]
  75. Tanaka, J.; Sugita, J.; Shiratori, S.; Shigematu, A.; Asanuma, S.; Fujimoto, K.; Nishio, M.; Kondo, T.; Imamura, M. Expansion of NK Cells from Cord Blood with Antileukemic Activity Using GMP-Compliant Substances without Feeder Cells. Leukemia 2012, 26, 1149–1152. [Google Scholar] [CrossRef]
  76. Granzin, M.; Stojanovic, A.; Miller, M.; Childs, R.; Huppert, V.; Cerwenka, A. Highly Efficient IL-21 and Feeder Cell-Driven Ex Vivo Expansion of Human NK Cells with Therapeutic Activity in a Xenograft Mouse Model of Melanoma. Oncoimmunology 2016, 5, e1219007. [Google Scholar] [CrossRef]
  77. Vasu, S.; Berg, M.; Davidson-Moncada, J.; Tian, X.; Cullis, H.; Childs, R.W. A Novel Method to Expand Large Numbers of CD56+ Natural Killer Cells from a Minute Fraction of Selectively Accessed Cryopreserved Cord Blood for Immunotherapy Post-Transplantation. Cytotherapy 2015, 17, 1582–1593. [Google Scholar] [CrossRef]
  78. Sakamoto, N.; Ishikawa, T.; Kokura, S.; Okayama, T.; Oka, K.; Ideno, M.; Sakai, F.; Kato, A.; Tanabe, M.; Enoki, T.; et al. Phase I Clinical Trial of Autologous NK Cell Therapy Using Novel Expansion Method in Patients with Advanced Digestive Cancer. J. Transl. Med. 2015, 13, 277. [Google Scholar] [CrossRef] [PubMed]
  79. Gong, W.; Xiao, W.; Hu, M.; Weng, X.; Qian, L.; Pan, X.; Ji, M. Ex Vivo Expansion of Natural Killer Cells with High Cytotoxicity by K562 Cells Modified to Co-Express Major Histocompatibility Complex Class I Chain-Related Protein A, 4-1BB Ligand, and Interleukin-15. Tissue Antigens 2010, 76, 467–475. [Google Scholar] [CrossRef] [PubMed]
  80. Fujisaki, H.; Kakuda, H.; Shimasaki, N.; Imai, C.; Ma, J.; Lockey, T.; Eldridge, P.; Leung, W.H.; Campana, D. Expansion of Highly Cytotoxic Human Natural Killer Cells for Cancer Cell Therapy. Cancer Res. 2009, 69, 4010–4017. [Google Scholar] [CrossRef]
  81. Ayello, J.; Hochberg, J.; Flower, A.; Chu, Y.; Baxi, L.V.; Quish, W.; van de Ven, C.; Cairo, M.S. Genetically Re-Engineered K562 Cells Significantly Expand and Functionally Activate Cord Blood Natural Killer Cells: Potential for Adoptive Cellular Immunotherapy. Exp. Hematol. 2017, 46, 38–47. [Google Scholar] [CrossRef]
  82. Denman, C.J.; Senyukov, V.V.; Somanchi, S.S.; Phatarpekar, P.V.; Kopp, L.M.; Johnson, J.L.; Singh, H.; Hurton, L.; Maiti, S.N.; Huls, M.H.; et al. Membrane-Bound IL-21 Promotes Sustained Ex Vivo Proliferation of Human Natural Killer Cells. PLoS ONE 2012, 7, e30264. [Google Scholar] [CrossRef]
  83. Yang, Y.; Badeti, S.; Tseng, H.C.; Ma, M.T.; Liu, T.; Jiang, J.G.; Liu, C.; Liu, D. Superior Expansion and Cytotoxicity of Human Primary NK and CAR-NK Cells from Various Sources via Enriched Metabolic Pathways. Mol. Ther. Methods Clin. Dev. 2020, 18, 428–445. [Google Scholar] [CrossRef]
  84. Kweon, S.; Phan, M.T.T.; Chun, S.; Yu, H.B.; Kim, J.; Kim, S.; Lee, J.; Ali, A.K.; Lee, S.H.; Kim, S.K.; et al. Expansion of Human NK Cells Using K562 Cells Expressing OX40 Ligand and Short Exposure to IL-21. Front. Immunol. 2019, 10, 879. [Google Scholar] [CrossRef]
  85. Guo, M.; Sun, C.; Qian, Y.; Zhu, L.; Ta, N.; Wang, G.; Zheng, J.; Guo, F.; Liu, Y. Proliferation of Highly Cytotoxic Human Natural Killer Cells by OX40L Armed NK-92 With Secretory Neoleukin-2/15 for Cancer Immunotherapy. Front. Oncol. 2021, 11, 632540. [Google Scholar] [CrossRef]
  86. Siegler, U.; Meyer-Monard, S.; Jrger, S.; Stern, M.; Tichelli, A.; Gratwohl, A.; Wodnar-Filipowicz, A.; Kalberer, C.P. Good Manufacturing Practice-Compliant Cell Sorting and Large-Scale Expansion of Single KIR-Positive Alloreactive Human Natural Killer Cells for Multiple Infusions to Leukemia Patients. Cytotherapy 2010, 12, 750–763. [Google Scholar] [CrossRef]
  87. Kim, E.; Ahn, Y.; Kim, S.; Kim, T.A.E.M.I.N.; Keam, B.; Heo, D.A.E.S. Ex Vivo Activation and Expansion of Natural Killer Cells from Patients with Advanced Cancer with Feeder Cells from Healthy Volunteers. J. Cytotherapy 2013, 15, 231–241.e1. [Google Scholar] [CrossRef]
  88. Delso-vallejo, M.; Kollet, J.; Koehl, U.; Huppert, V. Influence of Irradiated Peripheral Blood Mononuclear Cells on Both Ex Vivo Proliferation of Human Natural Killer Cells and Change in Cellular Property. Front. Immunol. 2017, 8, 854. [Google Scholar] [CrossRef] [PubMed]
  89. Yang, Y.; Lundqvist, A. Immunomodulatory Effects of IL-2 and IL-15; Implications for Cancer Immunotherapy. Cancers 2020, 12, 3586. [Google Scholar] [CrossRef] [PubMed]
  90. Isvoranu, G.; Surcel, M.; Munteanu, A.; Bratu, O.; Ionita-Radu, F.; Neagu, M.; Chiritoiu-Butnaru, M. Therapeutic Potential of Interleukin-15 in Cancer. Exp. Ther. Med. 2021, 22, 675. [Google Scholar] [CrossRef] [PubMed]
  91. Liao, W.; Lin, J.X.; Leonard, W.J. Interleukin-2 at the Crossroads of Effector Responses, Tolerance, and Immunotherapy. Immunity 2013, 38, 13–25. [Google Scholar] [CrossRef]
  92. Fehniger, T.A.; Cooper, M.A.; Caligiuri, M.A. Interleukin-2 and Interleukin-15 Immunotherapy for Cancer. Cytokine Growth Factor Rev. 2002, 13, 169–183. [Google Scholar] [CrossRef]
  93. Lehmann, D.; Spanholtz, J.; Sturtzel, C.; Tordoir, M.; Schlechta, B.; Groenewegen, D.; Hofer, E. IL-12 Directs Further Maturation of Ex Vivo Differentiated NK Cells with Improved Therapeutic Potential. PLoS ONE 2014, 9, e87131. [Google Scholar] [CrossRef]
  94. Hodge, D.L.; Subleski, J.J.; Reynolds, D.A.; Buschman, M.D.; Schill, W.B.; Burkett, M.W.; Malyguine, A.M.; Young, H.A. The Proinflammatory Cytokine Interleukin-18 Alters Multiple Signaling Pathways to Inhibit Natural Killer Cell Death. J. Interferon Cytokine Res. 2006, 26, 706–718. [Google Scholar] [CrossRef] [PubMed]
  95. Skak, K.; Frederiksen, K.S.; Lundsgaard, D. Interleukin-21 Activates Human Natural Killer Cells and Modulates Their Surface Receptor Expression. Immunology 2007, 123, 575–583. [Google Scholar] [CrossRef] [PubMed]
  96. Peighambarzadeh, F.; Najafalizadeh, A.; Esmaeil, N.; Rezaei, A.; Ashrafi, F.; Hakemi, M.G. Optimization of in Vitro Expansion and Activation of Human Natural Killer Cells against Breast Cancer Cell Line. Avicenna J. Med. Biotechnol. 2020, 12, 17–23. [Google Scholar] [PubMed]
  97. Haroun-Izquierdo, A.; Vincenti, M.; Netskar, H.; Van Ooijen, H.; Zhang, B.; Bendzick, L.; Kanaya, M.; Momayyezi, P.; Li, S.; Wiiger, M.T.; et al. Adaptive Single-KIR + NKG2C + NK Cells Expanded from Select Superdonors Show Potent Missing-Self Reactivity and Efficiently Control HLA-Mismatched Acute Myeloid Leukemia. J. Immunother. Cancer 2022, 10, e005577. [Google Scholar] [CrossRef] [PubMed]
  98. Liu, L.L.; Béziat, V.; Oei, V.Y.S.; Pfefferle, A.; Schaffer, M.; Lehmann, S.; Hellström-Lindberg, E.; Söderhäll, S.; Heyman, M.; Grandér, D.; et al. Ex Vivo Expanded Adaptive NK Cells Effectively Kill Primary Acute Lymphoblastic Leukemia Cells. Cancer Immunol. Res. 2017, 5, 654–665. [Google Scholar] [CrossRef]
  99. Condiotti, R.; Zakai, Y.B.; Barak, V.; Nagler, A. Ex Vivo Expansion of CD56 Cytotoxic Cells from Human Umbilical Cord Blood. Exp. Hematol. 2001, 29, 104–113. [Google Scholar] [CrossRef]
  100. Meehan, K.R.; Wu, J.; Webber, S.M.; Barber, A.; Szczepiorkowski, Z.M.; Sentman, C. Development of a Clinical Model for Ex Vivo Expansion of Multiple Populations of Effector Cells for Adoptive Cellular Therapy. Cytotherapy 2008, 10, 30–37. [Google Scholar] [CrossRef]
  101. Sutlu, T.; Stellan, B.; Gilljam, M.; Quezada, H.C.; Nahi, H.; Gahrton, G.Ö.S.T.A.; Alici, E. Clinical-Grade, Large-Scale, Feeder-Free Expansion of Highly Active Human Natural Killer Cells for Adoptive Immunotherapy Using an Automated Bioreactor. Cytotherapy 2010, 12, 1044–1055. [Google Scholar] [CrossRef] [PubMed]
  102. Lapteva, N.; Durett, A.G.; Sun, J.; Rollins, L.A.; Huye, L.L.; Fang, J.; Dandekar, V.; Mei, Z.; Jackson, K.; Vera, J.; et al. Large-Scale Ex Vivo Expansion and Characterization of Natural Killer Cells for Clinical Applications. Cytotherapy 2012, 14, 1131–1143. [Google Scholar] [CrossRef] [PubMed]
  103. Zwirner, N.W.; Domaica, C.I.; Fuertes, M.B. Regulatory Functions of NK Cells during Infections and Cancer. J. Leukoc. Biol. 2020, 109, 185–194. [Google Scholar] [CrossRef] [PubMed]
  104. Somanchi, S.S.; Senyukov, V.V.; Denman, C.J.; Lee, D.A. Expansion, Purification, and Functional Assessment of Human Peripheral Blood NK Cells. J. Vis. Exp. 2010, e2540. [Google Scholar] [CrossRef]
  105. Streltsova, M.A.; Ustiuzhanina, M.O.; Barsov, E.V.; Kust, S.A.; Velichinskii, R.A.; Kovalenko, E.I. Telomerase Reverse Transcriptase Increases Proliferation and Lifespan of Human NK Cells without Immortalization. Biomedicines 2021, 9, 662. [Google Scholar] [CrossRef] [PubMed]
  106. Fujisaki, H.; Kakuda, H.; Imai, C.; Mullighan, C.G.; Campana, D. Replicative Potential of Human Natural Killer Cells. Br. J. Haematol. 2009, 145, 606–613. [Google Scholar] [CrossRef]
  107. Konnikova, L.; Simeone, M.C.; Kruger, M.M.; Kotecki, M.; Cochran, B.H. Signal Transducer and Activator of Transcription 3 (STAT3) Regulates Human Telomerase Reverse Transcriptase (HTERT) Expression in Human Cancer and Primary Cells. Cancer Res. 2005, 65, 6516–6520. [Google Scholar] [CrossRef] [PubMed]
  108. Gurney, M.; Kundu, S.; Pandey, S.; O’Dwyer, M. Feeder Cells at the Interface of Natural Killer Cell Activation, Expansion and Gene Editing. Front. Immunol. 2022, 13, 802906. [Google Scholar] [CrossRef]
  109. Wang, K.S.; Frank, D.A.; Ritz, J. Interleukin-2 Enhances the Response of Natural Killer Cells to Interleukin-12 through up-Regulation of the Interleukin-12 Receptor and STAT4. Blood 2000, 95, 3183–3190. [Google Scholar] [CrossRef]
  110. Heipertz, E.L.; Zynda, E.R.; Stav-Noraas, T.E.; Hungler, A.D.; Boucher, S.E.; Kaur, N.; Vemuri, M.C. Current Perspectives on “Off-The-Shelf” Allogeneic NK and CAR-NK Cell Therapies. Front. Immunol. 2021, 12, 732135. [Google Scholar] [CrossRef]
  111. Maia, A.; Tarannum, M.; Romee, R. Genetic Manipulation Approaches to Enhance the Clinical Application of NK Cell-Based Immunotherapy. Stem Cells Transl. Med. 2023, szad087. [Google Scholar] [CrossRef]
  112. Maia, A.; Tarannum, M.; Romee, R. Cytokine-Induced Memory-Like NK Cells for Improved Cancer Immunotherapy. ASHI Q. Second Quart. 2023, 47, 30–34. [Google Scholar]
  113. Wang, S.; Sun, J.; Chen, K.; Ma, P.; Lei, Q.; Xing, S.; Cao, Z.; Sun, S.; Yu, Z.; Liu, Y.; et al. Perspectives of Tumor-Infiltrating Lymphocyte Treatment in Solid Tumors. BMC Med. 2021, 19, 140. [Google Scholar] [CrossRef]
  114. Tarannum, M.; Romee, R. Cytokine-Induced Memory-like Natural Killer Cells for Cancer Immunotherapy. Stem Cell Res. Ther. 2021, 12, 592. [Google Scholar] [CrossRef] [PubMed]
  115. Malone, D.F.G.; Lunemann, S.; Hengst, J.; Ljunggren, H.G.; Manns, M.P.; Sandberg, J.K.; Cornberg, M.; Wedemeyer, H.; Björkström, N.K. Cytomegalovirus-Driven Adaptive-like Natural Killer Cell Expansions Are Unaffected by Concurrent Chronic Hepatitis Virus Infections. Front. Immunol. 2017, 8, 525. [Google Scholar] [CrossRef] [PubMed]
  116. Stary, V.; Stary, G. NK Cell-Mediated Recall Responses: Memory-Like, Adaptive, or Antigen-Specific? Front. Cell. Infect. Microbiol. 2020, 10, 208. [Google Scholar] [CrossRef] [PubMed]
  117. Terrén, I.; Orrantia, A.; Mosteiro, A.; Vitallé, J.; Zenarruzabeitia, O.; Borrego, F. Metabolic Changes of Interleukin-12/15/18-Stimulated Human NK Cells. Sci. Rep. 2021, 11, 6472. [Google Scholar] [CrossRef] [PubMed]
  118. Lee, S.-H.; Fragoso, M.F.; Biron, C.A. Cutting Edge: A Novel Mechanism Bridging Innate and Adaptive Immunity: IL-12 Induction of CD25 To Form High-Affinity IL-2 Receptors on NK Cells. J. Immunol. 2012, 189, 2712–2716. [Google Scholar] [CrossRef] [PubMed]
  119. Romee, R.; Schneider, S.E.; Leong, J.W.; Chase, J.M.; Keppel, C.R.; Sullivan, R.P.; Cooper, M.A.; Fehniger, T.A. Cytokine Activation Induces Human Memory-like NK Cells. Blood 2012, 120, 4751–4760. [Google Scholar] [CrossRef] [PubMed]
  120. Cooper, M.A.; Elliott, J.M.; Keyel, P.A.; Yang, L.; Carrero, J.A.; Yokoyama, W.M. Cytokine-Induced Memory-like Natural Killer Cells. Proc. Natl. Acad. Sci. USA 2009, 106, 1915–1919. [Google Scholar] [CrossRef]
  121. Uppendahl, L.D.; Felices, M.; Bendzick, L.; Ryan, C.; Kodal, B.; Hinderlie, P.; Boylan, K.L.M.; Skubitz, A.P.N.; Miller, J.S.; Geller, M.A. Cytokine-Induced Memory-like Natural Killer Cells Have Enhanced Function, Proliferation, and In Vivo Expansion against Ovarian Cancer Cells. Gynecol. Oncol. 2019, 153, 149–157. [Google Scholar] [CrossRef]
  122. Romee, R.; Rosario, M.; Berrien-Elliott, M.M.; Wagner, J.A.; Jewell, B.A.; Schappe, T.; Leong, J.W.; Abdel-Latif, S.; Schneider, S.E.; Willey, S.; et al. Cytokine-Induced Memory-like Natural Killer Cells Exhibit Enhanced Responses against Myeloid Leukemia. Sci. Transl. Med. 2016, 8, 357ra123. [Google Scholar] [CrossRef] [PubMed]
  123. Barnes, S.; Schilizzi, O.; Audsley, K.M.; Newnes, H.V.; Foley, B. Deciphering the Immunological Phenomenon of Adaptive Natural Killer Cells and Cytomegalovirus. Int. J. Mol. Sci. 2020, 21, 8864. [Google Scholar] [CrossRef]
  124. Gumá, M.; Angulo, A.; Vilches, C.; Gómez-Lozano, N.; Malats, N.; López-Botet, M. Imprint of Human Cytomegalovirus Infection on the NK Cell Receptor Repertoire. Blood 2004, 104, 3664–3671. [Google Scholar] [CrossRef]
  125. Fesnak, A.D.; June, C.H.; Levine, B.L. Engineered T Cells: The Promise and Challenges of Cancer Immunotherapy. Nat. Rev. Cancer 2016, 16, 566–581. [Google Scholar] [CrossRef] [PubMed]
  126. Porter, D.L.; Levine, B.L.; Kalos, M.; Bagg, A.; June, C.H. Chimeric Antigen Receptor–Modified T Cells in Chronic Lymphoid Leukemia. N. Engl. J. Med. 2011, 365, 725–733. [Google Scholar] [CrossRef] [PubMed]
  127. Mikkilineni, L.; Kochenderfer, J.N. CAR T Cell Therapies for Patients with Multiple Myeloma. Nat. Rev. Clin. Oncol. 2021, 18, 71–84. [Google Scholar] [CrossRef]
  128. Pan, K.; Farrukh, H.; Chittepu, V.C.S.R.; Xu, H.; Pan, C.-X.; Zhu, Z. CAR Race to Cancer Immunotherapy: From CAR T, CAR NK to CAR Macrophage Therapy. J. Exp. Clin. Cancer Res. 2022, 41, 119. [Google Scholar] [CrossRef]
  129. Gang, M.; Marin, N.D.; Wong, P.; Neal, C.C.; Marsala, L.; Foster, M.; Schappe, T.; Meng, W.; Tran, J.; Schaettler, M.; et al. CAR-Modified Memory-like NK Cells Exhibit Potent Responses to NK-Resistant Lymphomas. Blood 2020, 136, 2308–2318. [Google Scholar] [CrossRef]
  130. Dong, H.; Ham, J.D.; Hu, G.; Xie, G.; Vergara, J.; Liang, Y.; Ali, A.; Tarannum, M.; Donner, H.; Baginska, J.; et al. Memory-like NK Cells Armed with a Neoepitope-Specific CAR Exhibit Potent Activity against NPM1 Mutated Acute Myeloid Leukemia. Proc. Natl. Acad. Sci. USA 2022, 119, e2122379119. [Google Scholar] [CrossRef]
  131. Daher, M.; Basar, R.; Gokdemir, E.; Baran, N.; Uprety, N.; Nunez Cortes, A.K.; Mendt, M.; Kerbauy, L.N.; Banerjee, P.P.; Shanley, M.; et al. Targeting a Cytokine Checkpoint Enhances the Fitness of Armored Cord Blood CAR-NK Cells. Blood 2021, 137, 624–636. [Google Scholar] [CrossRef]
  132. Zhu, H.; Blum, R.H.; Bernareggi, D.; Ask, E.H.; Wu, Z.; Hoel, H.J.; Meng, Z.; Wu, C.; Guan, K.L.; Malmberg, K.J.; et al. Metabolic Reprograming via Deletion of CISH in Human IPSC-Derived NK Cells Promotes In Vivo Persistence and Enhances Anti-Tumor Activity. Cell Stem Cell 2020, 27, 224–237.e6. [Google Scholar] [CrossRef]
  133. Bernard, P.L.; Delconte, R.; Pastor, S.; Laletin, V.; Costa Da Silva, C.; Goubard, A.; Josselin, E.; Castellano, R.; Krug, A.; Vernerey, J.; et al. Targeting CISH Enhances Natural Cytotoxicity Receptor Signaling and Reduces NK Cell Exhaustion to Improve Solid Tumor Immunity. J. Immunother. Cancer 2022, 10, e004244. [Google Scholar] [CrossRef]
  134. Shaim, H.; Shanley, M.; Basar, R.; Daher, M.; Gumin, J.; Zamler, D.B.; Uprety, N.; Wang, F.; Huang, Y.; Gabrusiewicz, K.; et al. Targeting the Av Integrin/TGF-β Axis Improves Natural Killer Cell Function against Glioblastoma Stem Cells. J. Clin. Investig. 2021, 131, e142116. [Google Scholar] [CrossRef]
  135. Kararoudi, M.; Nagai, Y.; Elmas, E.; Pereira, M.; Ali, S.; Imus, P.; Wethington, D.; Borrello, I.; Lee, D.; Ghiaur, G. CD38 Deletion of Human Primary NK Cells Eliminates Daratumumab-Induced Fratricide and Boosts Their Effector Activity. Blood 2020, 136, 2416–2427. [Google Scholar] [CrossRef]
  136. Ng, Y.Y.; Tay, J.C.K.; Wang, S. CXCR1 Expression to Improve Anti-Cancer Efficacy of Intravenously Injected CAR-NK Cells in Mice with Peritoneal Xenografts. Mol. Ther. Oncolytics 2020, 16, 75–85. [Google Scholar] [CrossRef]
  137. Ng, Y.Y.; Du, Z.; Zhang, X.; Chng, W.J.; Wang, S. CXCR4 and Anti-BCMA CAR Co-Modified Natural Killer Cells Suppress Multiple Myeloma Progression in a Xenograft Mouse Model. Cancer Gene Ther. 2022, 29, 475–483. [Google Scholar] [CrossRef] [PubMed]
  138. Schomer, N.T.; Jiang, Z.K.; Lloyd, M.I.; Klingemann, H.; Boissel, L. CCR7 Expression in CD19 Chimeric Antigen Receptor-Engineered Natural Killer Cells Improves Migration toward CCL19-Expressing Lymphoma Cells and Increases Tumor Control in Mice with Human Lymphoma. Cytotherapy 2022, 24, 827–834. [Google Scholar] [CrossRef] [PubMed]
  139. Brummel, K.; Eerkens, A.L.; de Bruyn, M.; Nijman, H.W. Tumour-Infiltrating Lymphocytes: From Prognosis to Treatment Selection. Br. J. Cancer 2023, 128, 451–458. [Google Scholar] [CrossRef] [PubMed]
  140. Dafni, U.; Michielin, O.; Lluesma, S.M.; Tsourti, Z.; Polydoropoulou, V.; Karlis, D.; Besser, M.J.; Haanen, J.; Svane, I.M.; Ohashi, P.S.; et al. Efficacy of Adoptive Therapy with Tumor-Infiltrating Lymphocytes and Recombinant Interleukin-2 in Advanced Cutaneous Melanoma: A Systematic Review and Meta-Analysis. Ann. Oncol. 2019, 30, 1902–1913. [Google Scholar] [CrossRef] [PubMed]
  141. Rosenberg, S.A.; Yannelli, J.R.; Yang, J.C.; Topalian, S.L.; Schwartzentruber, D.J.; Weber, J.S.; Parkinson, D.R.; Seipp, C.A.; Einhorn, J.H.; White, D.E. Treatment of Patients with Metastatic Melanoma with Autologous Tumor-Infiltrating Lymphocytes and Interleukin 2. J. Natl. Cancer Cent. 1994, 86, 1159–1166. [Google Scholar] [CrossRef]
  142. Chandran, S.S.; Somerville, R.P.T.; Yang, J.C.; Sherry, R.M.; Klebanoff, C.A.; Goff, S.L.; Wunderlich, J.R.; Danforth, D.N.; Zlott, D.; Paria, B.C.; et al. Treatment of Metastatic Uveal Melanoma with Adoptive Transfer of Tumour-Infiltrating Lymphocytes: A Single-Centre, Two-Stage, Single-Arm, Phase 2 Study. Lancet Oncol. 2017, 18, 792–802. [Google Scholar] [CrossRef]
  143. Zacharakis, N.; Chinnasamy, H.; Black, M.; Xu, H.; Lu, Y.C.; Zheng, Z.; Pasetto, A.; Langhan, M.; Shelton, T.; Prickett, T.; et al. Immune Recognition of Somatic Mutations Leading to Complete Durable Regression in Metastatic Breast Cancer. Nat. Med. 2018, 24, 724–730. [Google Scholar] [CrossRef]
  144. Zhao, Y.; Deng, J.; Rao, S.; Guo, S.; Shen, J.; Du, F.; Wu, X.; Chen, Y.; Li, M.; Chen, M.; et al. Tumor Infiltrating Lymphocyte (TIL) Therapy for Solid Tumor Treatment: Progressions and Challenges. Cancers 2022, 14, 4160. [Google Scholar] [CrossRef] [PubMed]
  145. Coca, S.; Perez-Piqueras, J.; Martinez, D.; Colmenarejo, A.; Saez, M.A.; Vallejo, C.; Martos, J.A.; Moreno, M. The Prognostic Significance of Intratumoral Natural Killer Cells in Patients with Colerectal Carcinoma. Cancer 1997, 79, 2320–2328. [Google Scholar] [CrossRef]
  146. Donadon, M.; Hudspeth, K.; Cimino, M.; Di Tommaso, L.; Preti, M.; Tentorio, P.; Roncalli, M.; Mavilio, D.; Torzilli, G. Increased Infiltration of Natural Killer and T Cells in Colorectal Liver Metastases Improves Patient Overall Survival. J. Gastrointest. Surg. 2017, 21, 1226–1236. [Google Scholar] [CrossRef]
  147. Ishigami, S.; Natsugoe, S.; Tokuda, K.; Nakajo, A.; Che, X.; Iwashige, H.; Aridome, K.; Hokita, S.; Aikou, T. Prognostic Value of Intratumoral Natural Killer Cells in Gastric Carcinoma. Cancer 2000, 88, 577–583. [Google Scholar] [CrossRef]
  148. Hoshikawa, M.; Aoki, T.; Matsushita, H.; Karasaki, T.; Hosoi, A.; Odaira, K.; Fujieda, N.; Kobayashi, Y.; Kambara, K.; Ohara, O.; et al. NK Cell and IFN Signatures Are Positive Prognostic Biomarkers for Resectable Pancreatic Cancer. Biochem. Biophys. Res. Commun. 2018, 495, 2058–2065. [Google Scholar] [CrossRef] [PubMed]
  149. Nersesian, S.; Schwartz, S.L.; Grantham, S.R.; MacLean, L.K.; Lee, S.N.; Pugh-Toole, M.; Boudreau, J.E. NK Cell Infiltration Is Associated with Improved Overall Survival in Solid Cancers: A Systematic Review and Meta-Analysis. Transl. Oncol. 2021, 14, 100930. [Google Scholar] [CrossRef] [PubMed]
  150. Cózar, B.; Greppi, M.; Carpentier, S.; Narni-Mancinelli, E.; Chiossone, L.; Vivier, E. Tumor-Infiltrating Natural Killer Cells. Cancer Discov. 2021, 11, 34–44. [Google Scholar] [CrossRef] [PubMed]
  151. Carrega, P.; Morandi, B.; Costa, R.; Frumento, G.; Forte, G.; Altavilla, G.; Ratto, G.B.; Mingari, M.C.; Moretta, L.; Ferlazzo, G. Natural Killer Cells Infiltrating Human Nonsmall-Cell Lung Cancer Are Enriched in CD56brightCD16 Cells and Display an Impaired Capability to Kill Tumor Cells. Cancer 2008, 112, 863–875. [Google Scholar] [CrossRef] [PubMed]
  152. Judge, S.J.; Darrow, M.A.; Thorpe, S.W.; Gingrich, A.A.; O’Donnell, E.F.; Bellini, A.R.; Sturgill, I.R.; Vick, L.V.; Dunai, C.; Stoffel, K.M.; et al. Analysis of Tumor-Infiltrating NK and T Cells Highlights IL-15 Stimulation and TIGIT Blockade as a Combination Immunotherapy Strategy for Soft Tissue Sarcomas. J. Immunother. Cancer 2020, 8, e001355. [Google Scholar] [CrossRef] [PubMed]
  153. Lim, S.A.; Kim, J.; Jeon, S.; Shin, M.H.; Kwon, J.; Kim, T.J.; Im, K.; Han, Y.; Kwon, W.; Kim, S.W.; et al. Defective Localization with Impaired Tumor Cytotoxicity Contributes to the Immune Escape of NK Cells in Pancreatic Cancer Patients. Front. Immunol. 2019, 10, 496. [Google Scholar] [CrossRef] [PubMed]
  154. Kremer, V.; Ligtenberg, M.; Zendehdel, R.; Seitz, C.; Duivenvoorden, A.; Wennerberg, E.; Colón, E.; Scherman-Plogell, A.H.; Lundqvist, A. Genetic Engineering of Human NK Cells to Express CXCR2 Improves Migration to Renal Cell Carcinoma. J. Immunother. Cancer 2017, 5, 73. [Google Scholar] [CrossRef]
  155. Giraldo, N.A.; Sanchez-Salas, R.; Peske, J.D.; Vano, Y.; Becht, E.; Petitprez, F.; Validire, P.; Ingels, A.; Cathelineau, X.; Fridman, W.H.; et al. The Clinical Role of the TME in Solid Cancer. Br. J. Cancer 2019, 120, 45–53. [Google Scholar] [CrossRef] [PubMed]
  156. Melaiu, O.; Lucarini, V.; Cifaldi, L.; Fruci, D. Influence of the Tumor Microenvironment on NK Cell Function in Solid Tumors. Front. Immunol. 2020, 10, 3038. [Google Scholar] [CrossRef] [PubMed]
  157. FDA. FDA Approves First Cellular Therapy to Treat Patients with Unresectable or Metastatic Melanoma; FDA: Silver Spring, MD, USA; Rockville, MD, USA, 2024.
  158. Mullard, A. FDA Approves First Tumour-Infiltrating Lymphocyte (TIL) Therapy, Bolstering Hopes for Cell Therapies in Solid Cancers. Nat. Rev. Drug Discov. 2024. [Google Scholar] [CrossRef]
  159. Liang, S.; Xu, K.; Niu, L.; Wang, X.; Liang, Y.; Zhang, M.; Chen, J.; Lin, M. Comparison of Autogeneic and Allogeneic Natural Killer Cells Immunotherapy on the Clinical Outcome of Recurrent Breast Cancer. OncoTargets Ther. 2017, 10, 4273–4281. [Google Scholar] [CrossRef]
  160. Yang, Y.; Lim, O.; Kim, T.M.; Ahn, Y.; Choi, H.; Chung, H.; Min, B.; Her, J.H.; Cho, S.Y.; Keam, B.; et al. Phase I Study of Random Healthy Donor—Derived Allogeneic Natural Killer Cell Therapy in Patients with Malignant Lymphoma or Advanced Solid Tumors. Cancer Immunol. Res. 2016, 4, 215–224. [Google Scholar] [CrossRef]
  161. Liu, S.; Galat, V.; Galat4, Y.; Lee, Y.K.A.; Wainwright, D.; Wu, J. NK Cell-Based Cancer Immunotherapy: From Basic Biology to Clinical Development. J. Hematol. Oncol. 2021, 14, 7. [Google Scholar] [CrossRef] [PubMed]
  162. Hermanson, D.L.; Bendzick, L.; Pribyl, L.; McCullar, V.; Vogel, R.I.; Miller, J.S.; Geller, M.A.; Kaufman, D.S. Induced Pluripotent Stem Cell-Derived Natural Killer Cells for Treatment of Ovarian Cancer. Stem Cells 2016, 34, 93–101. [Google Scholar] [CrossRef]
  163. Lee, S.C.; Shimasaki, N.; Lim, J.S.J.; Wong, A.; Yadav, K.; Yong, W.P.; Tan, L.K.; Koh, L.P.; Poon, M.L.M.; Tan, S.H.; et al. Phase I Trial of Expanded, Activated Autologous NK-Cell Infusions with Trastuzumab in Patients with HER2-Positive Cancers. Clin. Cancer Res. 2020, 26, 4494–4502. [Google Scholar] [CrossRef]
  164. Lim, C.M.; Liou, A.; Poon, M.; Koh, L.P.; Tan, L.K.; Loh, K.S.; Petersson, B.F.; Ting, E.; Campana, D.; Goh, B.C.; et al. Phase I Study of Expanded Natural Killer Cells in Combination with Cetuximab for Recurrent/Metastatic Nasopharyngeal Carcinoma. Cancer Immunol. Immunother. 2022, 71, 2277–2286. [Google Scholar] [CrossRef]
  165. Nahi, H.; Chrobok, M.; Meinke, S.; Gran, C.; Marquardt, N.; Afram, G.; Sutlu, T.; Gilljam, M.; Stellan, B.; Wagner, A.K.; et al. Autologous NK Cells as Consolidation Therapy Following Stem Cell Transplantation in Multiple Myeloma. Cell Rep. Med. 2022, 3, 100508. [Google Scholar] [CrossRef]
  166. Parkhurst, M.R.; Riley, J.P.; Dudley, M.E.; Rosenberg, S.A. Adoptive Transfer of Autologous Natural Killer Cells Leads to High Levels of Circulating Natural Killer Cells but Does Not Mediate Tumor Regression. Clin. Cancer Res. 2011, 17, 6287–6297. [Google Scholar] [CrossRef]
  167. Chu, J.; Gao, F.; Yan, M.; Zhao, S.; Yan, Z.; Shi, B.; Liu, Y. Natural Killer Cells: A Promising Immunotherapy for Cancer. J. Transl. Med. 2022, 20, 240. [Google Scholar] [CrossRef]
  168. Ruggeri, L.; Capanni, M.; Urbani, E.; Perruccio, K.; Shlomchik, W.D.; Tosti, A.; Posati, S.; Rogaia, D.; Frassoni, F.; Aversa, F.; et al. Effectiveness of Donor Natural Killer Cell Alloreactivity in Mismatched Hematopoietic Transplants. Science 2002, 295, 2097–2100. [Google Scholar] [CrossRef]
  169. Miller, J.S.; Cooley, S.; Parham, P.; Farag, S.S.; Verneris, M.R.; Mcqueen, K.L.; Guethlein, L.A.; Trachtenberg, E.A.; Haagenson, M.; Horowitz, M.M.; et al. Missing KIR Ligands Are Associated with Less Relapse and Increased Graft-versus-Host Disease (GVHD) Following Unrelated Donor Allogeneic HCT. Blood 2007, 109, 5058–5061. [Google Scholar] [CrossRef] [PubMed]
  170. Miller, J.S.; Soignier, Y.; Panoskaltsis-Mortari, A.; McNearney, S.A.; Yun, G.H.; Fautsch, S.K.; McKenna, D.; Le, C.; Defor, T.E.; Burns, L.J.; et al. Successful Adoptive Transfer and in Vivo Expansion of Human Haploidentical NK Cells in Patients with Cancer. Blood 2005, 105, 3051–3057. [Google Scholar] [CrossRef] [PubMed]
  171. Bachanova, V.; Sarhan, D.; Defor, T.; Cooley, S.; Panoskaltsis-, A.; Blazar, B.R.; Curtsinger, J.; Burns, L.; Weisdorf, D.J.; Miller, S.; et al. Haploidentical Natural Killer Cells Induce Remissions in Non-Hodgkin Lymphoma Patients With Low Levels of Immune-Suppressor Cells. Cancer Immunol. Immunother. 2018, 67, 483–494. [Google Scholar] [CrossRef] [PubMed]
  172. Bachanova, V.; Cooley, S.; Defor, T.E.; Verneris, M.R.; Zhang, B.; Mckenna, D.H.; Curtsinger, J.; Panoskaltsis-Mortari, A.; Lewis, D.; Hippen, K.; et al. Clearance of Acute Myeloid Leukemia by Haploidentical Natural Killer Cells Is Improved Using IL-2 Diphtheria Toxin Fusion Protein. Blood 2014, 123, 3855–3863. [Google Scholar] [CrossRef]
  173. Cooley, S.; He, F.; Bachanova, V.; Vercellotti, G.M.; DeFor, T.E.; Curtsinger, J.M.; Robertson, P.; Grzywacz, B.; Conlon, K.C.; Waldmann, T.A.; et al. First-in-Human Trial of RhIL-15 and Haploidentical Natural Killer Cell Therapy for Advanced Acute Myeloid Leukemia. Blood Adv. 2019, 3, 1970–1980. [Google Scholar] [CrossRef] [PubMed]
  174. Romee, R.; Cooley, S.; Berrien-Elliott, M.M.; Westervelt, P.; Verneris, M.R.; Wagner, J.E.; Weisdorf, D.J.; Blazar, B.R.; Ustun, C.; Defor, T.E.; et al. First-in-Human Phase 1 Clinical Study of the IL-15 Superagonist Complex ALT-803 to Treat Relapse after Transplantation. Blood 2018, 131, 2515–2527. [Google Scholar] [CrossRef] [PubMed]
  175. Shapiro, R.M.; Birch, G.C.; Hu, G.; Vergara Cadavid, J.; Nikiforow, S.; Baginska, J.; Ali, A.K.; Tarannum, M.; Sheffer, M.; Abdulhamid, Y.Z.; et al. Expansion, Persistence, and Efficacy of Donor Memory-like NK Cells Infused for Posttransplant Relapse. J. Clin. Investig. 2022, 132, e154334. [Google Scholar] [CrossRef] [PubMed]
  176. Shah, N.; Li, L.; McCarty, J.; Kaur, I.; Yvon, E.; Shaim, H.; Muftuoglu, M.; Liu, E.; Orlowski, R.Z.; Cooper, L.; et al. Phase I Study of Cord Blood-Derived Natural Killer Cells Combined with Autologous Stem Cell Transplantation in Multiple Myeloma. Br. J. Haematol. 2017, 177, 457–466. [Google Scholar] [CrossRef]
  177. Hoogstad-van Evert, J.; Bekkers, R.; Ottevanger, N.; Schaap, N.; Hobo, W.; Jansen, J.H.; Massuger, L.; Dolstra, H. Intraperitoneal Infusion of Ex Vivo-Cultured Allogeneic NK Cells in Recurrent Ovarian Carcinoma Patients (a Phase I Study). Medicine 2019, 98, e14290. [Google Scholar] [CrossRef]
  178. Dolstra, H.; Roeven, M.W.H.; Spanholtz, J.; Hangalapura, B.N.; Tordoir, M.; Maas, F.; Leenders, M.; Bohme, F.; Kok, N.; Trilsbeek, C.; et al. Successful Transfer of Umbilical Cord Blood CD34+ Hematopoietic Stem and Progenitor-Derived NK Cells in Older Acute Myeloid Leukemia Patients. Clin. Cancer Res. 2017, 23, 4107–4118. [Google Scholar] [CrossRef]
  179. Burga, R.A.; Nguyen, T.; Zulovich, J.; Madonna, S.; Ylisastigui, L.; Fernandes, R.; Yvon, E. Improving Efficacy of Cancer Immunotherapy by Genetic Modification of Natural Killer Cells. Cytotherapy 2016, 18, 1410–1421. [Google Scholar] [CrossRef]
  180. Cichocki, F.; Miller, J.S. In Vitro Development of Human Killer-Immunoglobulin Receptor-Positive NK Cells. Methods Mol. Biol. 2010, 612, 15–26. [Google Scholar] [CrossRef]
  181. Rafei, H.; Daher, M.; Katayoun, R. Chimeric Antigen Receptor (CAR) Natural Killer (NK)-Cell Therapy: Leveraging the Power of Innate Immunity. Br. J. Haematol. 2020, 193, 216–230. [Google Scholar] [CrossRef]
  182. Zhang, Z.; Wang, T.; Wang, X.; Zhang, Y.; Song, S.; Ma, C. Improving the Ability of CAR-T Cells to Hit Solid Tumors: Challenges and Strategies. Pharmacol. Res. 2021, 175, 106036. [Google Scholar] [CrossRef] [PubMed]
  183. Yeku, O.; Li, X.; Brentjens, R.J. Adoptive T-Cell Therapy for Solid Tumors. Am. Soc. Clin. Oncol. Educ. Book 2021, 37, 193–204. [Google Scholar] [CrossRef]
  184. Tong, L.; Jiménez-Cortegana, C.; Tay, A.H.M.; Wickström, S.; Galluzzi, L.; Lundqvist, A. NK Cells and Solid Tumors: Therapeutic Potential and Persisting Obstacles. Mol. Cancer 2022, 21, 206. [Google Scholar] [CrossRef] [PubMed]
  185. Rafiq, S.; Hackett, C.S.; Brentjens, R.J. Engineering Strategies to Overcome the Current Roadblocks in CAR T Cell Therapy. Nat. Rev. Clin. Oncol. 2020, 17, 147–167. [Google Scholar] [CrossRef] [PubMed]
  186. Xin, T.; Cheng, L.; Zhou, C.; Zhao, Y.; Hu, Z.; Wu, X. In-Vivo Induced CAR-T Cell for the Potential Breakthrough to Overcome the Barriers of Current CAR-T Cell Therapy. Front. Oncol. 2022, 12, 809754. [Google Scholar] [CrossRef]
  187. Hamilton, J.R.; Chen, E.; Perez, B.S.; Sandoval Espinoza, C.R.; Kang, M.H.; Trinidad, M.; Ngo, W.; Doudna, J.A. In Vivo Human T Cell Engineering with Enveloped Delivery Vehicles. Nat. Biotechnol. 2024. [Google Scholar] [CrossRef]
Figure 1. Representation of the NK cell subsets and NK-cell-mediated immunomodulation and cytotoxicity mechanisms. (A) NK cells are classified by CD56 and CD16 surface markers, including regulatory or immature NK cells (CD56bright CD16+/−), primarily releasing IFN-γ molecules, and cytotoxic or mature NK cells (CD56dim CD16+), mainly releasing perforin and granzyme B molecules. (B) Activated NK cells, in particular regulatory NK cells, release immunomodulatory mediators such as CCL5, XCL1, IL-8, IFN-γ, TNF-α, and GM-CSF within the TME, recruiting and activating other immune cells, including neutrophils, macrophages, dendritic cells (DC), T-cells, and B-cells. (C) Upon activation, NK cells exert cytotoxic effects through FasL and TRAIL-induced apoptosis, degranulation of perforin and granzyme molecules, and through antibody-dependent cell cytotoxicity (ADCC) via CD16, particularly cytotoxic NK cells, binding to antibodies on tumor cells, inducing apoptosis.
Figure 1. Representation of the NK cell subsets and NK-cell-mediated immunomodulation and cytotoxicity mechanisms. (A) NK cells are classified by CD56 and CD16 surface markers, including regulatory or immature NK cells (CD56bright CD16+/−), primarily releasing IFN-γ molecules, and cytotoxic or mature NK cells (CD56dim CD16+), mainly releasing perforin and granzyme B molecules. (B) Activated NK cells, in particular regulatory NK cells, release immunomodulatory mediators such as CCL5, XCL1, IL-8, IFN-γ, TNF-α, and GM-CSF within the TME, recruiting and activating other immune cells, including neutrophils, macrophages, dendritic cells (DC), T-cells, and B-cells. (C) Upon activation, NK cells exert cytotoxic effects through FasL and TRAIL-induced apoptosis, degranulation of perforin and granzyme molecules, and through antibody-dependent cell cytotoxicity (ADCC) via CD16, particularly cytotoxic NK cells, binding to antibodies on tumor cells, inducing apoptosis.
Cells 13 00451 g001aCells 13 00451 g001b
Figure 2. NK cell activation mechanisms. A balance of inhibitory and activating signals regulates NK cells. This balance is maintained when NK cells encounter normal cells, resulting in NK cell inactivity. However, certain conditions can activate NK cells, such as recognizing “missing self” when MHC-I molecules, serving as negative signals, are absent. Additionally, NK cells can be activated through “stress-induced” mechanisms when stress ligands, acting as positive signals, are overexpressed in stressed cells, including tumor- or viral-infected cells.
Figure 2. NK cell activation mechanisms. A balance of inhibitory and activating signals regulates NK cells. This balance is maintained when NK cells encounter normal cells, resulting in NK cell inactivity. However, certain conditions can activate NK cells, such as recognizing “missing self” when MHC-I molecules, serving as negative signals, are absent. Additionally, NK cells can be activated through “stress-induced” mechanisms when stress ligands, acting as positive signals, are overexpressed in stressed cells, including tumor- or viral-infected cells.
Cells 13 00451 g002
Table 1. Analysis of NK cell expansion rates across diverse cytokine-based expansion methods, varying in source, media, duration, and culture material.
Table 1. Analysis of NK cell expansion rates across diverse cytokine-based expansion methods, varying in source, media, duration, and culture material.
Ref.Cell SourceCulture FactorsTime (Days)Culture MaterialResults
[71]PBMCCellGro SCGM with 5% HS, anti-CD3 Ab for the first 5 days with 500 U/mL of IL-2.216-well plates and T25 flasks.CD3 CD56+ cells reached 193-fold expansion.
[72]PBMCSerum-free medium with 700 IU/mL IL-2, 0.01 KE/mL OK432, 10% human plasma, and an anti-CD16 Ab.21Flask and culture bag.Fold expansion ranges from 637 to 5712, with a purity of 76.9%.
[73]Isolated NK cellsX-VIVO 10 media with 5% heat-inactivated human FFP and 1000 U/mL of rhIL-2.12GMP-grade VueLife culture bags.The mean expansion rate of NK cells was 4-fold, while two donors reached 30-fold.
[74]Isolated NK cellsX-VIVO 10 media with 5% heat-inactivated human FFP, P/S, 100 or 1000 U/mL of IL-2, 10 ng/mL of IL-15, 25 ng/mL of IL-21 or combinations of those.42T25 flasks.IL-15 induced NK cell expansion, while IL-21 triggered NK cell maturation and functionality.
[75]CB-NK cellsSCGM with 5% human AB serum, IL-15, IL-2, anti-CD3 ab, tacrolimus, and dalteparin sodium.2024-well plates and T25 flasks.1700-fold expansion with 72.8% purity.
Disclaimer/Publisher’s Note: The statements, opinions and data contained in all publications are solely those of the individual author(s) and contributor(s) and not of MDPI and/or the editor(s). MDPI and/or the editor(s) disclaim responsibility for any injury to people or property resulting from any ideas, methods, instructions or products referred to in the content.

Share and Cite

MDPI and ACS Style

Maia, A.; Tarannum, M.; Lérias, J.R.; Piccinelli, S.; Borrego, L.M.; Maeurer, M.; Romee, R.; Castillo-Martin, M. Building a Better Defense: Expanding and Improving Natural Killer Cells for Adoptive Cell Therapy. Cells 2024, 13, 451. https://doi.org/10.3390/cells13050451

AMA Style

Maia A, Tarannum M, Lérias JR, Piccinelli S, Borrego LM, Maeurer M, Romee R, Castillo-Martin M. Building a Better Defense: Expanding and Improving Natural Killer Cells for Adoptive Cell Therapy. Cells. 2024; 13(5):451. https://doi.org/10.3390/cells13050451

Chicago/Turabian Style

Maia, Andreia, Mubin Tarannum, Joana R. Lérias, Sara Piccinelli, Luis Miguel Borrego, Markus Maeurer, Rizwan Romee, and Mireia Castillo-Martin. 2024. "Building a Better Defense: Expanding and Improving Natural Killer Cells for Adoptive Cell Therapy" Cells 13, no. 5: 451. https://doi.org/10.3390/cells13050451

Note that from the first issue of 2016, this journal uses article numbers instead of page numbers. See further details here.

Article Metrics

Back to TopTop