Preprint
Review

This version is not peer-reviewed.

Supercharged NK Cells Are a Unique Population of Cells Based on Transcriptomic, Single-Cell RNAseq, Proteomic, and Functional Characteristics; Comparison with Other NK Cell Subpopulations

A peer-reviewed article of this preprint also exists.

Submitted:

11 June 2025

Posted:

12 June 2025

You are already at the latest version

Abstract
Natural killer (NK) cells are large granular lymphocytes with potent anti-tumor activity. However, their use in clinical trials has resulted in less than favorable outcomes due to the lack of reliable methods to augment their expansion and anti-tumor potential. Based on the previous publications and our ongoing studies, we present the characteristics of sNK cells that make these cells optimal for clinical trial applications. sNK cells have significantly high anti-tumor activity as well as increased survival and augmented expansion when they are cultured with osteoclasts and probiotic bacteria. In this review, we compare the expansion and function of sNK cells with other NK cell expansion methodologies to indicate the similarities and differences between sNK cells and other NK cell expansion methodologies. We also describe the uniqueness of sNK cells in terms of increased survival, expansion, and heightened function as determined by proteomic, transcriptomic, single-cell RNAseq, and functional characteristics. These cells not only target and kill poorly differentiated/stem-like tumors, but also can target differentiated tumors. In addition, they can restore the functional loss of autologous NK cells, allowing the patient's NK cells to become functional. These attributes of sNK cells make these cells not only unique population of NK cells but also cells that can effectively be used in clinical trials of cancer patients.
Keywords: 
;  ;  ;  ;  ;  

1. Why Do We Need NK Cell-Based Cancer Immunotherapies?

Current treatments for the most aggressive tumors include surgery, radiation therapy, chemotherapy (examples include but are not limited to Paclitaxel, Gemcitabine, Fluorouracil, Irinotecan liposome injection, etc.), targeted therapy, T-cell therapy, and other forms of immunotherapy. In advanced cases, these treatments are not likely to offer a benefit; thus, the physician will focus on palliative care for as long as possible [1,2]. Most explored cancer immunotherapies include: monoclonal antibodies, checkpoint inhibitors, bispecific antibodies, T Cell redirecting antibodies, vaccination strategies, gene-modified T cells, donor lymphocyte infusions (DLI) and antigen-specific DLI, CAR T-cells, and TCR gene-modified T cells. Interest in adoptive cell therapy for treating cancer is exploding, given the early clinical successes of autologous chimeric antigen receptor (CAR) T lymphocyte therapy [3,4,5]. However, limitations using T cells and autologous cell products are apparent as they (A) take weeks to generate, (B) utilize a 1:1 donor-to-patient model, (C) are expensive, and (D) are prone to heterogeneity and manufacturing failures [6]. CAR T cells are also associated with significant toxicities, including cytokine release syndrome, immune effector cell–associated neurotoxicity syndrome, and prolonged cytopenia [7,8]. To overcome these issues, natural killer cells (NK) are being explored as an alternative cell source for allogeneic cell therapies [9,10,11], and are currently being investigated in ongoing clinical trials as single therapy or combination therapy [12,13,14,15], and have thus far yielded many encouraging clinical results [16].
NK cells may represent a valid alternative to T cells, due to their inherent nature as part of the innate immune response to aggressively attack major histocompatibility complex (MHC) class I-deficient or mutated cells [17]. Although tumors may develop multiple resistance mechanisms to endogenous NK cell attack, in vitro activation, expansion, and genetic modification of NK cells can greatly enhance their anti-tumor activity and give them the ability to overcome drug resistance [18,19]. Unlike T cells, NK cells activation is managed by the interaction of NK cells receptors with their ligands on the target cell; this process is not dependent on antigen processing or presentation [20]. NK cells can be obtained from blood or cord blood, or be derived from hematopoietic stem and progenitor cells or induced pluripotent stem cells, and can be expanded and cryopreserved for off-the-shelf availability [20,21,22]. NK cell progenitors or mature NK cells can be infused with other cells as part of the Hematopoietic stem-cell transplantation (HSCT) or alone following the pre-enrichment process. Inhibitory receptors on donor allogeneic NK cells (e.g., KIR) do not recognize human leukocyte antigen (HLA) class I on recipient cells in case of a class mismatch. Therefore, the donor NK cells are relieved of their repressive receptor-triggered inhibition. In this case, cancer cells lack the suitable class I MHC ligands to engage the repressive KIR, and thus, they are removed by allo-reactive NK cells [16,23,24]. Numerous reports have revealed that allogeneic NK cells potentially trigger remission or suppress relapse of the tumor in cancer patients [25,26]. In a clinical trial of haploidentical NK cells for AML, the authors reported the induction of complete remission in dismal prognosis or elderly individuals and a 100% event-free survival rate at 18 months in a pediatric cohort [1,2]. Through these advances, NK cell–based therapies provide a complementary clinical strategy to, and overcome limitations of, US Food and Drug Administration–approved chimeric antigen receptor T-cell therapies [27,28,29].
Natural killer (NK) cells are the large granular Lymphocytes with potent anti-tumor and anti-viral functions. However, clinical use of these cells is hampered due to a lack of reliable methods to augment their expansion and anti-tumor potential. Based on the previous publications, in this review, we summarize the characteristics of supercharged NK (sNK) cells that make these cells optimal to be used in cell therapy. We provide a comprehensive review on sNK, the NK cell subsets with significantly increased anti-cancer activity (cytotoxic function and the secretion levels of cytokines). The technology to develop supercharged NK cells was demonstrated in several publications generated from our laboratory [30,31,32,33]. We also discuss the preclinical efficacy and safety of sNK cells in humanized mice [31,32,34,35,36,37,38] and clinical efficacy and safety in human patients infused with sNK cells [3][].

2. Supercharged NK Cells

To generate highly potent supercharged NK cells (sNK), PBMC-derived NK cells are co-cultured with osteoclasts (OCs) and sonicated probiotic bacteria (sAJ2). Osteoclasts were selected among several feeder cells because they express ligands for NK cell receptors such as MICA/B, KLRG1, and ULBPs on their surface and secrete cytokines and chemokines, including IL-12, IL-15, IFN-γ, and IL-18, which are known to activate NK cells [40,41]. Probiotic bacteria sAJ2 is a combination of seven gram-positive probiotic bacteria Streptococcus thermophiles, Bifidobacterium longum, Bifidobacterium breve, Bifidobacterium infantis, Lactobacillus acidophilus, Lactobacillus plantarum, and Lactobacillus paracasei. These strains were selected based on their activation in NK cells. The combination of these probiotic treatments in NK cells was found to increase cytokine secretion by NK cells, including IFN-γ, which could facilitate signals required for NK cell expansion [42,43,44,45,46]. Therefore, a combination treatment of both probiotics and OCs in NK cells results in the induction of signals participating in cell expansion and functional activation of NK cells, generating sNK cells. sNK cells have demonstrated increased life-span, cell expansion, cytotoxicity, and secretion of cytokines; these sNK cell characteristics ultimately result in increased differentiation and killing of oral cancer both in vivo and in vitro [32,35,44].
sNK cells are a unique population of NK cells with completely distinct profiles from those of primary peripheral blood-derived NK cells at the RNA seq analysis at the single cell level based on UMAP (an algorithm that takes a high-dimensional dataset) and regulon profiles, and in terms of cell cycle analysis, granule content, and functional capabilities [31]. sNK cells have very high anti-tumor cytotoxicity and are polyfunctional [31]. sNK cells demonstrated higher expression of activating receptors and down-modulation of inhibitory receptors [44]. In addition, sNK cells demonstrated increased secretion of IFN-γ and TNF-α, elevated levels of Trail expression at the single-cell transcriptomic analysis, higher levels of BCL2, and were able to resist the induction of cell death and loss of cytotoxicity within the tumor microenvironment [47]. Moreover, in contrast to primary activated NK cells, sNK cells were able to lyse both stem-like and differentiated tumors [48]. sNK cells were found to be superior to many other NK cell treatments, including IL-2, IL-2 and anti-CD16 mAbs, IL-2 and anti-CD16 mAbs, and sAJ2 [47,49], treatment with other NK-specific cytokines such as IL-12, IL-15, and IL-18 and OSCSCs or K562 expanded NK cells [44,47,50,51]. In addition, when compared to cord blood-derived NK cells, iPSC-derived NK cells, NK92, and several other NK cells, sNK cells exhibited much higher levels of cytotoxicity and cytokine secretion [36,48]. sNK cells induce tumor killing irrespective of MHC-class I expression on tumor cells, making them a unique NK cell-based treatment strategy in heterogeneous tumors [36]. We recently explored the proteomic, transcriptomic, and functional characterization of sNK cells and found that sNK cells were less susceptible to split anergy and tumor-induced exhaustion. Split anergy is the stage of NK cells previously defined in our studies [52,53,54]. At this stage, NK cells reduced their cytotoxic function in the presence of significant secretion of cytokines [52,53,54]. Proteomic analyses revealed that sNK cells significantly increase their cell motility and proliferation. Single-cell transcriptomes indicated that sNK cells undertake a unique differentiation trajectory and turn on several important regulons essential for augmenting anti-tumor effector functions and proliferation. Both proteomic and single-cell transcriptomes revealed that an increase in Cathepsin C augmented the quantity and function of Granzyme B. The results obtained support the use of sNK cells for clinical utilization and delineate the molecular mechanisms associated with their maturation [31,36,55] (manuscript in press).

3. History: A Historical View of NK Expansion Methodologies and Their Differences with Supercharged NK Cells

Since its discovery, the NK biologists have attempted to discover strategies that could be safely used to effectively treat cancer patients. Even though these cells were shown to be extremely safe in cell therapy, the level of efficacy has always been challenged due to several issues. Unlike T cells, which can comprise 60 to 40% of lymphocytes, NK cells are only 5-10% on average in the peripheral blood [56]. Also, the functions of NK cells are impacted during preneoplasia and cancer [57,58]. NK cells are quick to become inactivated in the tumor microenvironment, even when they are activated by cytokines or other methods. There is a lack of persistence of NK cells when injected into the patients. These qualities of NK cells have made these cells less desirable for cell therapy. However, recently, several methodologies have been found to overcome the lack of adequate expansion of NK cells ex vivo [30,59,60,61,62,63,64,65,66,67]. Unfortunately, many of such methodologies give rise to larger numbers of NK cells with poor quality of for a methodology to not only expand well but also retain NK cell functional capabilities [56]. Even though we are getting closer to having effective cells, we are still not entirely there. Although many studies claim that they have a significant expansion of NK cells with the use of cytokines, the majority of them have been less desirable due to the variability of the donor-derived NK cells' expansion and functional capabilities. Thus, the search for super donors makes NK cell-based therapy very limited. Feeder layer-dependent expansion, such as K562 or OSCSCs, has been less than desirable for expansion [30,31]. Also, genetic manipulation expressing 4IBBL and IL-15 or IL-21 has gained popularity for the expansion of the NK cells [30,65,68,69,70,71,72,73,74,75]. It is not clear whether such expansion will increase the functional activation of NK cells maximally and for how long. Knockdown of genes in NK cells was shown to increase the functional activation of NK cells [76]. This characteristic is very common for the function of NK cells since either the knockdown is in the target cells or the NK cells will result in the activation of the NK cells [76,77]. Again, no experiments have been performed to compare this platform to the other existing expansion methodologies; as such, it is difficult to compare with other platforms.
Although we did not have access to all the different kinds of platforms that have been engineered for expansion and functional activation of NK cells, we had the opportunity to compare the sNK cells with a few established strategies. When comparing sNK cells to either IL-2-activated cord blood-derived NK cells or iPSC-derived NK cells, no NK cell-mediated cytotoxicity and slight IFN-γ secretion could be seen, whereas there were substantial increases in cytotoxicity and secretion of IFN-γ by the sNK cells. We have characterized NK92 cells for their function previously and found them to have no or very slight cytotoxicity and secretion of IFN-γ [48]. In addition, using K562 or OSCSCs as feeder cells, we could not see appreciable levels of expansion and function [30,31]. Likewise, although we could see an increase in NK cell function with the combination of cytokine treatment, we could not see increased expansion of the cells [30,31]. When we used K562 or OSCSCs or MP2 or PBMCs as feeder cells to expand NK cells, and compared to NK expanded using OCs as feeder cells, an increased rate of NK cell proliferation, increased NK cell survival in culture, much higher cytotoxicity, and higher levels of cytokine secretions were seen in NK cells expanded using OCs as feeder cells [30,31].

4. Current Findings:

4.1. Infusion of sNK Cells in Humanized Mice or Humans Leads to a Significant Increase in the Percentages of NK Cells and Restores or Increases Function in Autologous NK Cells

When assessing the expansion and function of autologous NK cells after sNK infusion, either in mice (31, 32, 34, 35) or humans (manuscript in press), we could see a substantial improvement in the NK function in pancreatic, oral, and melanoma-implanted humanized mice [31,32,34,35]. There was a 47% to 94% improvement in NK cytotoxicity depending on the tissue examined when sNK cells were infused in pancreatic tumor-bearing mice (Table 1A). Likewise, there was 69% to 97% improvement in secretion of IFN-γ, depending on the tissue examined, when sNK cells were infused in pancreatic tumor-bearing mice (Table 1A). Similarly, there was 78% to 98% improvement in NK cytotoxicity depending on the tissue examined when sNK cells were infused in oral cancer-bearing mice (Table 1B). There was 76% to 98% improvement in secretion of IFN-γ, depending on the tissue examined, when sNK cells were infused in oral cancer-bearing mice (Table 1B). There was 69% to 85% improvement in NK cytotoxicity, depending on the tissue examined, when sNK cells were infused in melanoma-bearing mice (Table 1C). There was a 56% to 93% improvement in the secretion of IFN-γ, depending on the tissue examined, when sNK cells were infused in melanoma-bearing mice (Table 1C). In humans, we observed increased percentages of autologous NK cells and increased function of NK cells in cancer patients (manuscript in press).
Table 1: Humanized mice were orthotopically injected with 1 x 10⁶ human MIA PaCa-2 (MP2) (A), oral tumor CSCs (B), or melanoma tumor cells (C) into the pancreas, oral, and subcutaneously, respectively. For pancreatis and oral tumors, one week post-tumor implantation, mice were administered 1 x 10⁶ sNK cells via tail-vein injection. For melanoma, one week post-tumor implantation, mice were administered 2 x 10⁶ sNK cells every 7 days via tail-vein injection. At week 5, the mice were sacrificed, and tissues were collected. Single-cell isolation from tissues, NK cells, or T cells were cultured with IL-2 (1000 U/mL) for 7 days. After incubation, cells were used as effectors against OSCSCs in a standard 4-hour ⁵¹Cr release assay (cytotoxicity), and the supernatants were collected to measure IFN-γ secretion levels using ELISA. The restoration of cytotoxic function and IFN-γ secretion was quantified using the formula:(LU or IFN-γ of sNK-treated tumor mice / LU or IFN-γ of healthy mice) × 100.

4.2. Safety of sNK Cells for Human Use

In terms of safety, there was no evidence of treatment-related toxicities or graft vs. host disease [31,32,34,35]. Indeed, when infused into humans, patients indicated that they felt more energy and all their aches and pains had disappeared. The safety of these cells was determined both in adult populations and in pediatric populations. Unlike sNK cell infusion, treatment-related toxicities were very frequently seen in other well-established cancer therapeutics, such as CAR-T therapies, which are found to be associated with cytokine-release syndrome (CRS) and immune effector cell-associated neurotoxicity syndrome (ICANS) [78,79,80,81,82,83,84]. In addition, in CAR-T or CAR-NK cells, if the tumor downmodulates or loses the particular CAR, it has to be manufactured again, whereas sNK cells target all different types of tumors. Unlike CART cells, sNK cells do not lead to cytokine-release syndrome (CRS) and immune effector cell-associated neurotoxicity syndrome. sNK cells do not undergo cell death while interacting with tumor cells, therefore, they have longer circulation times and more effective function. sNK cells are easy to scale up, and we can obtain greater than a billion sNK cells for therapy. Finally, sNK cells can be used with other therapeutics to further enhance the eradication of tumors. Since sNK cells are known to expand CD8+ T cells, the combination of sNK with CD8+ T cells will result in better tumor clearance. Finally, sNK cells can target both poorly differentiated/stem-like tumors and the differentiated tumors [36]. These findings make sNK cells an effective cell therapeutic strategy to eliminate tumors and hopefully achieve a cure for the patients.
These data indicate that sNK cells not only can target and eliminate tumors, but they can also increase the percentages and function of autologous NK cells. This increase was observed greater than 9 months in cancer patients.

4. Conclusions

The methodology to expand sNK cells does not involve any genetic manipulation of cells. sNK cells work differently than CART or CARNK cells since they can target tumors with/without a wide range of antigens, on a wide variety of cancers. sNK cells can target both CSCs as well as differentiated tumors, facilitating the targeting of the heterogeneous population of tumors, ultimately leading to complete eradication of cancer. sNK cells secrete significantly higher levels of cytokines, especially those known to induce differentiation of tumors. sNK-differentiated tumors can be further cleared by CD8+ T cells or chemotherapy. Thus, sNK cells could enhance the efficacy of other therapies. sNK cells induce beneficial effects with no signs of adverse events, particularly they don’t induce cytokine-release syndrome (CRS) or immune effector cell-associated neurotoxicity syndrome. Restored or increased immune function post-sNK cell therapy in patients results in an immune system boost so that autologous NK cells can further fight against the disease. Last, but not least, it is easy to scale up the sNK cells’ production for cancer therapeutics as the starting material is the immune cells from the peripheral blood of a healthy donor, which are easily available.

Author Contributions

KK performed the data generation, literature search, and wrote the manuscript. AJ was the principal investigator on research articles cited in this review, and she also participated in writing and editing the manuscript.

Conflicts of Interest

The authors declare no conflict of interest.

References

  1. Stathopoulos, G.P.; Dimitroulis, J.; Antoniou, D.; Katis, C.; Tsavdaridis, D.; Armenaki, O.; et al. Front-line paclitaxel and irinotecan combination chemotherapy in advanced non-small-cell lung cancer: a phase I-II trial. Br J Cancer. 2005, 93, 1106–1111. [Google Scholar] [CrossRef] [PubMed]
  2. Sochacka-Ćwikła, A.; Mączyński, M.; Regiec, A. FDA-Approved Drugs for Hematological Malignancies-The Last Decade Review. Cancers (Basel). 2021, 14. [Google Scholar] [CrossRef] [PubMed]
  3. June, C.H.; O'Connor, R.S.; Kawalekar, O.U.; Ghassemi, S.; Milone, M.C. CAR T cell immunotherapy for human cancer. Science. 2018, 359, 1361–1365. [Google Scholar] [CrossRef]
  4. Maude, S.L.; Frey, N.; Shaw, P.A.; Aplenc, R.; Barrett, D.M.; Bunin, N.J.; et al. Chimeric antigen receptor T cells for sustained remissions in leukemia. N Engl J Med. 2014, 371, 1507–1517. [Google Scholar] [CrossRef] [PubMed]
  5. Levine, B.L.; Miskin, J.; Wonnacott, K.; Keir, C. Global Manufacturing of CAR T Cell Therapy. Mol Ther Methods Clin Dev. 2017, 4, 92–101. [Google Scholar] [CrossRef]
  6. Xiao, X.; Huang, S.; Chen, S.; Wang, Y.; Sun, Q.; Xu, X.; et al. Mechanisms of cytokine release syndrome and neurotoxicity of CAR T-cell therapy and associated prevention and management strategies. J Exp Clin Cancer Res. 2021, 40, 367. [Google Scholar] [CrossRef]
  7. Siegler, E.L.; Kenderian, S.S. Neurotoxicity and Cytokine Release Syndrome After Chimeric Antigen Receptor T Cell Therapy: Insights Into Mechanisms and Novel Therapies. Front Immunol. 2020, 11, 1973. [Google Scholar] [CrossRef]
  8. Srivastava, S.; Riddell, S.R. Chimeric Antigen Receptor T Cell Therapy: Challenges to Bench-to-Bedside Efficacy. J Immunol. 2018, 200, 459–468. [Google Scholar] [CrossRef]
  9. Li, Y.; Rezvani, K.; Rafei, H. Next-generation chimeric antigen receptors for T- and natural killer-cell therapies against cancer. Immunol Rev. 2023, 320, 217–235. [Google Scholar] [CrossRef]
  10. Zhang, Y.; Xu, Y.; Dang, X.; Zhu, Z.; Qian, W.; Liang, A.; et al. Challenges and optimal strategies of CAR T therapy for hematological malignancies. Chin Med J (Engl). 2023, 136, 269–279. [Google Scholar] [CrossRef]
  11. Bridgeman, J.S.; Hawkins, R.E.; Hombach, A.A.; Abken, H.; Gilham, D.E. Building better chimeric antigen receptors for adoptive T cell therapy. Curr Gene Ther. 2010, 10, 77–90. [Google Scholar] [CrossRef] [PubMed]
  12. Noh, J.Y.; Seo, H.; Lee, J.; Jung, H. Immunotherapy in Hematologic Malignancies: Emerging Therapies and Novel Approaches. Int J Mol Sci. 2020, 21. [Google Scholar] [CrossRef] [PubMed]
  13. Bachireddy, P.; Burkhardt, U.E.; Rajasagi, M.; Wu, C.J. Haematological malignancies: at the forefront of immunotherapeutic innovation. Nat Rev Cancer. 2015, 15, 201–215. [Google Scholar] [CrossRef] [PubMed]
  14. Peng, L.; Sferruzza, G.; Yang, L.; Zhou, L.; Chen, S. CAR-T and CAR-NK as cellular cancer immunotherapy for solid tumors. Cell Mol Immunol. 2024, 21, 1089–1108. [Google Scholar] [CrossRef]
  15. Yang, R.; Yang, Y.; Liu, R.; Wang, Y.; Yang, R.; He, A. Advances in CAR-NK cell therapy for hematological malignancies. Front Immunol. 2024, 15, 1414264. [Google Scholar] [CrossRef]
  16. Berrien-Elliott, M.M.; Jacobs, M.T.; Fehniger, T.A. Allogeneic natural killer cell therapy. Blood. 2023, 141, 856–868. [Google Scholar] [CrossRef]
  17. Daher, M.; Rezvani, K. Next generation natural killer cells for cancer immunotherapy: the promise of genetic engineering. Curr Opin Immunol. 2018, 51, 146–53. [Google Scholar] [CrossRef]
  18. Page, A.; Chuvin, N.; Valladeau-Guilemond, J.; Depil, S. Development of NK cell-based cancer immunotherapies through receptor engineering. Cell Mol Immunol. 2024, 21, 315–331. [Google Scholar] [CrossRef]
  19. Sordo-Bahamonde, C.; Vitale, M.; Lorenzo-Herrero, S.; López-Soto, A.; Gonzalez, S. Mechanisms of Resistance to NK Cell Immunotherapy. Cancers (Basel). 2020, 12. [Google Scholar] [CrossRef]
  20. Liu, S.; Galat, V.; Galat, 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]
  21. Fang, F.; Wang, W.; Chen, M.; Tian, Z.; Xiao, W. Technical advances in NK cell-based cellular immunotherapy. Cancer Biol Med. 2019, 16, 647–654. [Google Scholar] [CrossRef]
  22. Maia, A.; Tarannum, M.; Lérias, J.R.; Piccinelli, S.; Borrego, L.M.; Maeurer, M.; et al. Building a Better Defense: Expanding and Improving Natural Killer Cells for Adoptive Cell Therapy. Cells. 2024, 13. [Google Scholar] [CrossRef]
  23. Gao, F.; Ye, Y.; Gao, Y.; Huang, H.; Zhao, Y. Influence of KIR and NK Cell Reconstitution in the Outcomes of Hematopoietic Stem Cell Transplantation. Front Immunol. 2020, 11, 2022. [Google Scholar] [CrossRef]
  24. Mehta, R.S.; Rezvani, K. Can we make a better match or mismatch with KIR genotyping? Hematology Am Soc Hematol Educ Program. 2016, 2016, 106–118. [Google Scholar] [CrossRef] [PubMed]
  25. Mohseni, R.; Mahdavi Sharif, P.; Behfar, M.; Shojaei, S.; Shoae-Hassani, A.; Jafari, L.; et al. Phase I study of safety and efficacy of allogeneic natural killer cell therapy in relapsed/refractory neuroblastomas post autologous hematopoietic stem cell transplantation. Sci Rep. 2024, 14, 20971. [Google Scholar] [CrossRef]
  26. Bednarski, J.J.; Zimmerman, C.; Berrien-Elliott, M.M.; Foltz, J.A.; Becker-Hapak, M.; Neal, C.C.; et al. Donor memory-like NK cells persist and induce remissions in pediatric patients with relapsed AML after transplant. Blood. 2022, 139, 1670–1683. [Google Scholar] [CrossRef]
  27. Heipertz, E.L.; Zynda, E.R.; Stav-Noraas, T.E.; Hungler, A.D.; Boucher, S.E.; Kaur, N.; et al. Current Perspectives on "Off-The-Shelf" Allogeneic NK and CAR-NK Cell Therapies. Front Immunol. 2021, 12, 732135. [Google Scholar] [CrossRef]
  28. Guillerey, C.; Huntington, N.D.; Smyth, M.J. Targeting natural killer cells in cancer immunotherapy. Nat Immunol. 2016, 17, 1025–1036. [Google Scholar] [CrossRef]
  29. Lamb, M.G.; Rangarajan, H.G.; Tullius, B.P.; Lee, D.A. Natural killer cell therapy for hematologic malignancies: successes, challenges, and the future. Stem Cell Res Ther. 2021, 12, 211. [Google Scholar] [CrossRef]
  30. Kaur, K.; Cook, J.; Park, S.-H.; Topchyan, P.; Kozlowska, A.; Ohanian, N.; et al. Novel Strategy to Expand Super-Charged NK Cells with Significant Potential to Lyse and Differentiate Cancer Stem Cells: Differences in NK Expansion and Function between Healthy and Cancer Patients. Frontiers in Immunology. 2017, 8, 2017. [Google Scholar] [CrossRef]
  31. Kaur, K.; Chen, P.-C.; Ko, M.-W.; Mei, A.; Senjor, E.; Malarkannan, S.; et al. Sequential therapy with supercharged NK cells with either chemotherapy drug cisplatin or anti-PD-1 antibody decreases the tumor size and significantly enhances the NK function in Hu-BLT mice. Frontiers in Immunology. 2023, 14, 2023. [Google Scholar] [CrossRef] [PubMed]
  32. Kaur, K.; Ko, M.-W.; Ohanian, N.; Cook, J.; Jewett, A. Osteoclast-expanded super-charged NK-cells preferentially select and expand CD8+ T cells. Scientific Reports. 2020, 10, 20363. [Google Scholar] [CrossRef] [PubMed]
  33. Chiang, J.; Chen, P.-C.; Pham, J.; Nguyen, C.-Q.; Kaur, K.; Raman, S.S.; et al. Characterizing hepatocellular carcinoma stem markers and their corresponding susceptibility to NK-cell based immunotherapy. Frontiers in Immunology. 2023, 14, 2023. [Google Scholar] [CrossRef] [PubMed]
  34. Kaur, K.; Paytsar, T.; Karolina, K.A.; Nick, O.; Jessica, C.; Ou, M.P.; et al. Super-charged NK cells inhibit growth and progression of stem-like/poorly differentiated oral tumors in vivo in humanized BLT mice; effect on tumor differentiation and response to chemotherapeutic drugs. OncoImmunology. 2018, 7, e1426518. [Google Scholar] [CrossRef]
  35. Kaur, K.; Kozlowska, A.K.; Topchyan, P.; Ko, M.-W.; Ohanian, N.; Chiang, J.; et al. Probiotic-Treated Super-Charged NK Cells Efficiently Clear Poorly Differentiated Pancreatic Tumors in Hu-BLT Mice. Cancers. 2020, 12, 63. [Google Scholar] [CrossRef]
  36. Huerta-Yepez, S.; Chen, P.C.; Kaur, K.; Jain, Y.; Singh, T.; Esedebe, F.; et al. Supercharged NK cells, unlike primary activated NK cells, effectively target ovarian cancer cells irrespective of MHC-class I expression. BMJ Oncol. 2025, 4, e000618. [Google Scholar] [CrossRef]
  37. Jewett, A.; Kaur, K.; Gharamanians, N. Supercharged NK Cells as a Promising Therapeutic Strategy to Target and Eliminate Aggressive DIPG Tumors in Pediatric Patients. Critical Reviews™ in Immunology 2025, 45, 13–16. [Google Scholar] [CrossRef]
  38. Kaur, K.; Jewett, A. Supercharged NK Cell-Based Immuotherapy in Humanized Bone Marrow Liver and Thymus (Hu-BLT) Mice Model of Oral, Pancreatic, Glioblastoma, Hepatic, Melanoma and Ovarian Cancers. 2023, 43, 13–25.
  39. Jewett, A. First in Human Clinical Study Demonstrating the Safety and Efficacy of NK101 (supercharged NK cells) in the Treatment of Patients with Cancer. South East European Journal of Immunology. 2025, 8, 040. [Google Scholar] [CrossRef]
  40. Tseng, H.C.; Kanayama, K.; Kaur, K.; Park, S.H.; Park, S.; Kozlowska, A.; et al. Bisphosphonate-induced differential modulation of immune cell function in gingiva and bone marrow in vivo: role in osteoclast-mediated NK cell activation. Oncotarget. 2015, 6, 20002–20025. [Google Scholar] [CrossRef]
  41. Li, H.; Hong, S.; Qian, J.; Zheng, Y.; Yang, J.; Yi, Q. Cross talk between the bone and immune systems: osteoclasts function as antigen-presenting cells and activate CD4+ and CD8+ T cells. Blood. 2010, 116, 210–217. [Google Scholar] [CrossRef]
  42. Bui, V.T.; Tseng, H.C.; Kozlowska, A.; Maung, P.O.; Kaur, K.; Topchyan, P.; et al. Augmented IFN-γ and TNF-α Induced by Probiotic Bacteria in NK Cells Mediate Differentiation of Stem-Like Tumors Leading to Inhibition of Tumor Growth and Reduction in Inflammatory Cytokine Release; Regulation by IL-10. Front Immunol. 2015, 6, 576. [Google Scholar] [CrossRef] [PubMed]
  43. Kaur, K.; Topchyan, P.; Kozlowska, A.K.; Ohanian, N.; Chiang, J.; Maung, P.O.; et al. Super-charged NK cells inhibit growth and progression of stem-like/poorly differentiated oral tumors in vivo in humanized BLT mice; effect on tumor differentiation and response to chemotherapeutic drugs. Oncoimmunology. 2018, 7, e1426518. [Google Scholar] [CrossRef]
  44. Kaur, K.; Cook, J.; Park, S.H.; Topchyan, P.; Kozlowska, A.; Ohanian, N.; et al. Novel Strategy to Expand Super-Charged NK Cells with Significant Potential to Lyse and Differentiate Cancer Stem Cells: Differences in NK Expansion and Function between Healthy and Cancer Patients. Front Immunol. 2017, 8, 297. [Google Scholar] [CrossRef]
  45. Kaur, K.; Kozlowska, A.K.; Topchyan, P.; Ko, M.W.; Ohanian, N.; Chiang, J.; et al. Probiotic-Treated Super-Charged NK Cells Efficiently Clear Poorly Differentiated Pancreatic Tumors in Hu-BLT Mice. Cancers (Basel). 2019, 12. [Google Scholar] [CrossRef] [PubMed]
  46. Dong, H.; Rowland, I.; Yaqoob, P. Comparative effects of six probiotic strains on immune function in vitro. Br J Nutr. 2012, 108, 459–470. [Google Scholar] [CrossRef]
  47. Kaur, K.; Chen, P.C.; Ko, M.W.; Mei, A.; Senjor, E.; Malarkannan, S.; et al. Sequential therapy with supercharged NK cells with either chemotherapy drug cisplatin or anti-PD-1 antibody decreases the tumor size and significantly enhances the NK function in Hu-BLT mice. Front Immunol. 2023, 14, 1132807. [Google Scholar] [CrossRef]
  48. Kaur, K.; Safaie, T.; Ko, M.-W.; Wang, Y.; Jewett, A. ADCC against MICA/B Is Mediated against Differentiated Oral and Pancreatic and Not Stem-Like/Poorly Differentiated Tumors by the NK Cells; Loss in Cancer Patients due to Down-Modulation of CD16 Receptor. Cancers. 2021, 13, 239. [Google Scholar] [CrossRef]
  49. Chiang, J.; Chen, P.-C.; Pham, J.; Nguyen, C.-Q.; Kaur, K.; Raman, S.S.; et al. Characterizing hepatocellular carcinoma stem markers and their corresponding susceptibility to NK-cell based immunotherapy. Frontiers in Immunology. 2023, 14. [Google Scholar] [CrossRef] [PubMed]
  50. Romee, R.; Rosario, M.; Berrien-Elliott, M.M.; Wagner, J.A.; Jewell, B.A.; Schappe, T.; et al. Cytokine-induced memory-like natural killer cells exhibit enhanced responses against myeloid leukemia. Sci Transl Med. 2016, 8, 357ra123. [Google Scholar] [CrossRef]
  51. Terrén, I.; Orrantia, A.; Astarloa-Pando, G.; Amarilla-Irusta, A.; Zenarruzabeitia, O.; Borrego, F. Cytokine-Induced Memory-Like NK Cells: From the Basics to Clinical Applications. Front Immunol. 2022, 13, 884648. [Google Scholar] [CrossRef]
  52. Magister, S.; Obermajer, N.; Mirkovic, B.; Svajger, U.; Renko, M.; Softic, A.; et al. Regulation of cathepsins S and L by cystatin F during maturation of dendritic cells. Eur J Cell Biol. 2012, 91, 391–401. [Google Scholar] [CrossRef] [PubMed]
  53. Tseng, H.C.; Bui, V.; Man, Y.G.; Cacalano, N.; Jewett, A. Induction of Split Anergy Conditions Natural Killer Cells to Promote Differentiation of Stem Cells through Cell-Cell Contact and Secreted Factors. Front Immunol. 2014, 5, 269. [Google Scholar] [CrossRef] [PubMed]
  54. Tseng, H.C.; Cacalano, N.; Jewett, A. Split anergized Natural Killer cells halt inflammation by inducing stem cell differentiation, resistance to NK cell cytotoxicity and prevention of cytokine and chemokine secretion. Oncotarget. 2015, 6, 8947–8959. [Google Scholar] [CrossRef]
  55. Kaur, K.; Jewett, A. Super-Charged Natural Killer Cells: A Promising Immunotherapeutic Strategy for Oral Cancer. Immuno. 2025, 5, 8. [Google Scholar] [CrossRef]
  56. Fang, F.; Xie, S.; Chen, M.; Li, Y.; Yue, J.; Ma, J.; et al. Advances in NK cell production. Cellular & Molecular Immunology. 2022, 19, 460–481. [Google Scholar]
  57. Kaur, K.; Chang, H.H.; Cook, J.; Eibl, G.; Jewett, A. Suppression of Gingival NK Cells in Precancerous and Cancerous Stages of Pancreatic Cancer in KC and BLT-Humanized Mice. Frontiers in immunology. 2017, 8, 1606. [Google Scholar] [CrossRef]
  58. Jewett, A.; Kos, J.; Kaur, K.; Safaei, T.; Sutanto, C.; Chen, W.; et al. Natural Killer Cells: Diverse Functions in Tumor Immunity and Defects in Pre-neoplastic and Neoplastic Stages of Tumorigenesis. Mol Ther Oncolytics. 2020, 16, 41–52. [Google Scholar] [CrossRef] [PubMed]
  59. Perussia, B.; Ramoni, C.; Anegon, I.; Cuturi, M.C.; Faust, J.; Trinchieri, G. Preferential proliferation of natural killer cells among peripheral blood mononuclear cells cocultured with B lymphoblastoid cell lines. Natural immunity and cell growth regulation. 1987, 6, 171–188. [Google Scholar]
  60. Rabinowich, H.; Sedlmayr, P.; Herberman, R.B.; Whiteside, T.L. Increased proliferation, lytic activity, and purity of human natural killer cells cocultured with mitogen-activated feeder cells. Cellular immunology. 1991, 135, 454–470. [Google Scholar] [CrossRef]
  61. Igarashi, T.; Wynberg, J.; Srinivasan, R.; Becknell, B.; McCoy, J.P.; Jr Takahashi, Y.; et al. Enhanced cytotoxicity of allogeneic NK cells with killer immunoglobulin-like receptor ligand incompatibility against melanoma and renal cell carcinoma cells. Blood. 2004, 104, 170–177. [Google Scholar] [CrossRef]
  62. Srivastava, S.; Lundqvist, A.; Childs, R.W. Natural killer cell immunotherapy for cancer: a new hope. Cytotherapy. 2008, 10, 775–783. [Google Scholar] [CrossRef]
  63. Gras Navarro, A.; Björklund, A.; Chekenya, M. Therapeutic potential and challenges of Natural killer cells in treatment of solid tumors. Frontiers in Immunology. 2015, 6. [Google Scholar] [CrossRef] [PubMed]
  64. Alici, E.; Sutlu, T.; Bjorkstrand, B.; Gilljam, M.; Stellan, B.; Nahi, H.; et al. Autologous antitumor activity by NK cells expanded from myeloma patients using GMP-compliant components. Blood. 2008, 111, 3155–3162. [Google Scholar] [CrossRef] [PubMed]
  65. Fujisaki, H.; Kakuda, H.; Shimasaki, N.; Imai, C.; Ma, J.; Lockey, T.; et al. Expansion of highly cytotoxic human natural killer cells for cancer cell therapy. Cancer research. 2009, 69, 4010–4017. [Google Scholar] [CrossRef] [PubMed]
  66. Berg, M.; Lundqvist, A.; McCoy, P.; Jr Samsel, L.; Fan, Y.; Tawab, A.; et al. Clinical-grade ex vivo-expanded human natural killer cells up-regulate activating receptors and death receptor ligands and have enhanced cytolytic activity against tumor cells. Cytotherapy. 2009, 11, 341–355. [Google Scholar] [CrossRef]
  67. Garg, T.K.; Szmania, S.M.; Khan, J.A.; Hoering, A.; Malbrough, P.A.; Moreno-Bost, A.; et al. Highly activated and expanded natural killer cells for multiple myeloma immunotherapy. Haematologica. 2012, 97, 1348–1356. [Google Scholar] [CrossRef]
  68. Voskens, C.J.; Watanabe, R.; Rollins, S.; Campana, D.; Hasumi, K.; Mann, D.L. Ex-vivo expanded human NK cells express activating receptors that mediate cytotoxicity of allogeneic and autologous cancer cell lines by direct recognition and antibody directed cellular cytotoxicity. Journal of experimental & clinical cancer research : CR. 2010, 29, 134. [Google Scholar]
  69. Yang, H.; Tang, R.; Li, J.; Liu, Y.; Ye, L.; Shao, D.; et al. A New Ex Vivo Method for Effective Expansion and Activation of Human Natural Killer Cells for Anti-Tumor Immunotherapy. Cell biochemistry and biophysics. 2015, 73, 723–729. [Google Scholar] [CrossRef]
  70. Fujisaki, H.; Kakuda, H.; Imai, C.; Mullighan, C.G.; Campana, D. Replicative potential of human natural killer cells. British journal of haematology. 2009, 145, 606–613. [Google Scholar] [CrossRef]
  71. Chang, Y.H.; Connolly, J.; Shimasaki, N.; Mimura, K.; Kono, K.; Campana, D. A chimeric receptor with NKG2D specificity enhances natural killer cell activation and killing of tumor cells. Cancer research. 2013, 73, 1777–1786. [Google Scholar] [CrossRef]
  72. Lapteva, N.; Durett, A.G.; Sun, J.; Rollins, L.A.; Huye, L.L.; Fang, 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]
  73. Moseman, J.E.; Foltz, J.A.; Sorathia, K.; Heipertz, E.L.; Lee, D.A. Evaluation of serum-free media formulations in feeder cell-stimulated expansion of natural killer cells. Cytotherapy. 2020, 22, 322–328. [Google Scholar] [CrossRef] [PubMed]
  74. Zhao, X.Y.; Jiang, Q.; Jiang, H.; Hu, L.J.; Zhao, T.; Yu, X.X.; et al. Expanded clinical-grade membrane-bound IL-21/4-1BBL NK cell products exhibit activity against acute myeloid leukemia in vivo. Eur J Immunol. 2020, 50, 1374–1385. [Google Scholar] [CrossRef]
  75. Zhang, C.; Kadu, S.; Xiao, Y.; Johnson, O.; Kelly, A.; O'Connor, R.S.; et al. Sequential Exposure to IL21 and IL15 During Human Natural Killer Cell Expansion Optimizes Yield and Function. Cancer Immunol Res. 2023, 11, 1524–1537. [Google Scholar] [CrossRef]
  76. Tseng, H.-C.; Arasteh, A.; Kaur, K.; Kozlowska, A.; Topchyan, P.; Jewett, A. Differential cytotoxicity but augmented IFN-γ secretion by NK cells after interaction with monocytes from humans, and those from wild type and myeloid specific COX-2 knockout mice. Frontiers in Immunology. 2015, 6, 2015. [Google Scholar] [CrossRef]
  77. Kozlowska, A.K.; Topchyan, P.; Kaur, K.; Tseng, H.C.; Teruel, A.; Hiraga, T.; et al. Differentiation by NK cells is a prerequisite for effective targeting of cancer stem cells/poorly differentiated tumors by chemopreventive and chemotherapeutic drugs. J Cancer. 2017, 8, 537–554. [Google Scholar] [CrossRef]
  78. Schultz, L.; Mackall, C. Driving CAR T cell translation forward. Sci Transl Med. 2019, 11. [Google Scholar] [CrossRef] [PubMed]
  79. Sterner, R.C.; Sterner, R.M. CAR-T cell therapy: current limitations and potential strategies. Blood Cancer J. 2021, 11, 69. [Google Scholar] [CrossRef]
  80. Neelapu, S.S.; Locke, F.L.; Bartlett, N.L.; Lekakis, L.J.; Miklos, D.B.; Jacobson, C.A.; 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]
  81. Park, J.H.; Rivière, I.; Gonen, M.; Wang, X.; Sénéchal, B.; Curran, K.J.; et al. Long-Term Follow-up of CD19 CAR Therapy in Acute Lymphoblastic Leukemia. N Engl J Med. 2018, 378, 449–459. [Google Scholar] [CrossRef]
  82. Schuster, S.J.; Bishop, M.R.; Tam, C.S.; Waller, E.K.; Borchmann, P.; McGuirk, J.P.; et al. Tisagenlecleucel in Adult Relapsed or Refractory Diffuse Large B-Cell Lymphoma. N Engl J Med. 2019, 380, 45–56. [Google Scholar] [CrossRef] [PubMed]
  83. Wudhikarn, K.; Pennisi, M.; Garcia-Recio, M.; Flynn, J.R.; Afuye, A.; Silverberg, M.L.; et al. DLBCL patients treated with CD19 CAR T cells experience a high burden of organ toxicities but low nonrelapse mortality. Blood Adv. 2020, 4, 3024–3033. [Google Scholar] [CrossRef] [PubMed]
  84. Morgan, R.A.; Yang, J.C.; Kitano, M.; Dudley, M.E.; Laurencot, C.M.; Rosenberg, S.A. Case report of a serious adverse event following the administration of T cells transduced with a chimeric antigen receptor recognizing ERBB2. Mol Ther. 2010, 18, 843–851. [Google Scholar] [CrossRef] [PubMed]
Table 1. Efficacy of sNK cells in restoring immune function in tumor-bearing humanized mice.
Table 1. Efficacy of sNK cells in restoring immune function in tumor-bearing humanized mice.
A. Pancreatic tumor-bearing humanized mice
Tissues or isolated cells P values (tumor untreated mice vs. tumor mice treated with sNK cells) (LU or IFN-γ of sNK cell-treated tumor mice/LU or IFN-γ of healthy mice)*100 # of mice
Cytotoxicity IFN-γ Cytotoxicity IFN-γ
Peripheral blood ** * 94% 69% 9
Bone marrow * * 75% 67% 9
Spleen * * 66% 72% 9
Pancreas/pancreatic tumor ** ** 88% 97% 9
Oral mucosa ** * 92% 84% 6
NK cells isolated from spleen * * 64% 59% 9
NK cells isolated from PBMCs * * 47% 69% 9
B. Oral tumor-bearing humanized mice
Tissues or isolated cells P values (tumor untreated mice vs. tumor mice treated with sNK cells) (LU or IFN-γ of sNK cell treated tumor mice/LU or IFN-γ of healthy mice)*100 # of mice
Cytotoxicity IFN-γ Cytotoxicity IFN-γ
Peripheral blood ** ** 98.00% 97% 8
Bone marrow ** ** 96.70% 98% 8
Spleen ** ** 92.80% 96% 8
Oral mucosa ** * 94% 87% 5
NK cells isolated from spleen * * 77.80% 76% 8
NK cells isolated from PBMCs ** *** 88% 98% 8
B. Melanoma-bearing humanized mice
Tissues or isolated cells P values (tumor untreated mice vs. tumor mice treated with sNK cells) (LU or IFN-γ of sNK cell-treated tumor mice/LU or IFN-γ of healthy mice)*100 # of mice
Cytotoxicity IFN-γ Cytotoxicity IFN-γ
Peripheral blood * * 73% 76% 6
Bone marrow * * 69% 77% 6
Spleen NA * NA 56% 6
NK cells isolated from the spleen ** ** 85% 88% 6
T cells isolated from PBMCs ** ** 83% 93% 6
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.
Copyright: This open access article is published under a Creative Commons CC BY 4.0 license, which permit the free download, distribution, and reuse, provided that the author and preprint are cited in any reuse.
Prerpints.org logo

Preprints.org is a free preprint server supported by MDPI in Basel, Switzerland.

Subscribe

Disclaimer

Terms of Use

Privacy Policy

Privacy Settings

© 2026 MDPI (Basel, Switzerland) unless otherwise stated