Skip to main content

OPINION article

Front. Oncol., 02 December 2022
Sec. Cancer Immunity and Immunotherapy
This article is part of the Research Topic NK cell modifications to advance their anti-tumor activities View all 13 articles

Overcoming the challenges in translational development of natural killer cell therapeutics: An opinion paper

Hong QinHong Qin1Changqiao You*Changqiao You1*Feng YanFeng Yan1Kefang TanKefang Tan1Changgen XuChanggen Xu1Rui ZhaoRui Zhao2Marlene Davis Ekpo*Marlene Davis Ekpo2*Songwen TanSongwen Tan2
  • 1Research and Development Department, NanHua Bio-medicine CO., Ltd., Changsha, China
  • 2Xiangya School of Pharmaceutical Sciences, Central South University, Changsha, China

Introduction

Cellular therapies have attracted huge research and clinical attention lately (1, 2). Natural killer cells (NKCs) are a class of innate immune lymphoid cells (ILP) mainly derived from bone marrow lymphoid stem cells (3). They are mainly distributed in peripheral blood (PB) and peripheral lymphoid tissues, accounting for about 10% of the total lymphocytes in PB (47). Presently, NK cell immunotherapeutics utilize cells derived from many sources including PB, umbilical cord blood (8), immortalized NK cell lines, and more recently, induced pluripotent stem cells (iPSC) (911). They are characterized by rapid response and non-specific cytotoxic effect without prior antigen sensitization, and are independent on antibodies or complements (12, 13). NKCs induce apoptosis of target cells by secreting perforin, granzyme, cytokines and chemokines (14, 15). NKCs also selectively attacks foreign and diseased cells through expression of killer-cell immunoglobulin-like receptors (KIRs) and Fc receptor (CD16) where the later mediates antibody-dependent cell-medicated cytotoxicity (ADCC) (1618).

Human NKCs are defined by the CD3-CD56+ surface phenotype (19, 20). According to their surface expression of CD56, they are divided into two main subpopulations: CD56bright and CD56dim, which differ significantly in biological characteristics (2022). Briefly, CD56bright NKCs are immature cells and the progenitor for effector cells with high expression of CD56 and low expression of CD16 and KIRs (21), and accounts for about 5- 10% of the total NKCs. Also, they are weakly immunoregulatory and rely mainly on the secretion of cytokines, growth factors and chemokines (22). On the contrary, CD56dim subset accounts for about 90-95% of the circulating NKCs. They are characterized by low expression of CD56 and high expression of CD16, KIRs, FcγRIII and a variety of NK cell inhibitory receptors. They exhibit intrinsic cytotoxicity and ADCC but have weak cytokine secretion ability (23, 24).

Many studies have revealed the role of NKCs in pathologies like autoimmune diseases (25, 26), leukemia (25), pregnancy-related conditions (27), liver diseases (2830), HIV (31), HPV (32), atherosclerosis (33) and in many age-related diseases. Aging is usually associated with increased susceptibility to infectious diseases and cancers due to immunosenescence which is particularly reflected in the biological changes in the population and subsets of NKCs throughout life (34). As the functioning of the immune system decreases with age, NKCs have difficulties in initiating adaptive immune responses and mobilizing effective immune molecules, leading to the occurrence of diseases related to aging, such as infections and tumors (3538). The proportion of CD56dim NKCs in the elderly increased with age, while the CD56bright NKCs decreased significantly, suggesting that the increased CD56dimNK : CD56brightNK ratio is significantly age related (39, 40). Studies have also shown that the mortality risk in the elderly with low NK cell counts is three-fold higher than that in the elderly with high NK cell counts (41). In addition to the decreased CD56bright NKCs in older adults (4244), Sagiv et al. demonstrated that the decline in perforin-mediated NK cytotoxicity is similarly age-related, and may hinder the ability of NKCs to clear senescent cells in the elderly (45). Similarly, NKCs from geriatric population have synonymous reduction in proliferative response to interleukin 2 (IL-2) and expression of the CD69 activation antigen (4649). With immunosenescence being almost inevitable, many are willing to explore therapies to escape the negative consequences of aging especially tumor development which is increasingly prevalent. Therefore, undergoing NK cell therapy in early old age may help particularly in alleviating cancers.

Studies have shown that the therapeutic efficiency of NK cell therapeutics while encouraging in hematopoietic malignancies, is unsatisfactory in solid tumors as it is problematic for NKCs to infiltrate tumor sites (50, 51). The function, activation, and persistence of NKCs are significantly diminished by the tumor microenvironment (TME), leading to their dysfunction or exhaustion. In this paper, we wish to draw the attention of researchers to the fact that NK cellular products are highly promising in the fight against cancer and other age-complicated diseases and if they must be applied safely, more efforts should be directed towards addressing the bottlenecks. Broadly, NK cell infiltration, solid tumor targeting, in vivo persistence and resistance to TME must be improved, and reproducible and standardized protocols must be developed for the generation and expansion of NKCs. Here in, we highlight the strategies employed in tackling the challenges as this will serve as guide to the research trend and future directions considered in the development of clinical grade NK biotherapeutics.

Improving tumor targeting, microenvironment resistance and solid tumor infiltration

Clinical trials involving NKCs is generally not a new topic as they have been in progress for over two decades from where preliminary data regarding the safety and efficacy of NKCs have been obtained following their adoptive transfer to treat hematologic malignancies (25, 52). Nevertheless, some limitations have been encountered in the application of NK cell therapy to solid tumors largely because the TME harbors suppressive ligands, metabolites and cytokines which threatens the survival of NKCs (53). Additionally, the tumor itself possesses other defense mechanisms against attack by NKCs. Therefore, enormous research efforts are directed towards producing or modifying NKCs to be more resistant to attack from tumors and TME while harnessing their cytolytic effect after tumor penetration (54, 55). One of such approaches include blocking of inhibitory receptors with monoclonal antibodies (mAbs) like monalizumab (56).

Increasing the efficacy of tumor cell recognition is achievable via genetic modification (57) (58). For example, CAR-NKCs have enhanced cytolytic activity attributable to the synergistic effect of targeted specificity against tumor associated antigens and intracellular signaling of receptors (59, 60). CAR-NKCs can also be fashioned with receptors for a wide range of antigens with the CAR expression permitting carrier cells to recognize antigens on tumor-cell surfaces without major histocompatibility complex restriction (9, 58, 61). Also, unlike CAR-T cell therapies, CAR-NKCs possess reduced risk of cytokine release syndrome, neurological complications and better potential for allogeneic applications (62).

Transfection efficiency for primary NKCs is a key obstacle to the large-scale manufacture of genetically modified CAR-NKCs and different techniques like viral transduction and non-viral electroporation are underway to addressing this challenge (63, 64). Kumar et al. has recently led the production and evaluation of CRISPR-engineered NK-92 cell constitutively expressing Cas9 or dCas9 which have shown good prospects for further research and possible clinical application (65).

Pharmacokinetics and pharmacodynamics of natural killer cell therapeutics

NK and other cellular therapeutics are different from conventional (chemical) drugs therefore great disparity exists in their pharmacokinetics and pharmacodynamics properties. There is substantial evidence from clinical studies regarding the safety and efficacy of NK cell therapeutics. Nonetheless, more research is on demand to explore the sensitivity a wider variety of tumors to NK cell therapy, determine the mechanism(s) of action (cytotoxic response) against different types of tumors and identify possible contraindications. Some of the identified issues are as thus:

1. Allogenic NKCs from PB are relatively safe and satisfactorily effective against tumors but are susceptible to rejection by the host (66, 67).

2. Although several studies show the relationship between high NKCs, their receptors/ligands levels and better overall survival in patients with hepatocellular carcinoma (HCC), the underlying mechanism of remains unclear (13).

3. In obese patients, significant numbers of NK and T cells are recruited to the visceral adipose tissue at the expense of successful tumor infiltration and eradication (68), thus posing a serious challenge for the application of NK cell therapy in certain comorbid situations.

4. Combination therapy with NK and T cells or other tumor therapy strategies need to be confirmed with large-scale clinical trials as the clinical outcome can vary between tumor types.

Hence, several research efforts are geared towards unravelling possible NK mechanism of cytolysis like mitochondrial apoptosis (64) and release of perforin and granzymes (69); factors that may increase their cytotoxicity such as E26 transformation-specific transcription factor ELK3 expression by cancer cells (70); factors that attenuate cytolytic function like increased transforming growth factor-beta 1 (TGF-β1) (32), inhibition of O-GlcNAcylation (71), low surface expression and impaired function of transient receptor potential melastatin 3 (TRPM3) (32, 7175); and addressing the complications accompanying rejection-prone cellular products (67, 76, 77).

Expansion and activation of natural killer cells

Another obstacle to the manufacture of clinical grade NK cell therapeutics is the large-scale expansion of NKCs without loss of their cytotoxic activity (78). The expansion of NKCs can be ex vivo or in vivo followed by isolation by CD3+ cell depletion and subsequent positive selection of CD56+ cells. Other strategies involve a single step depletion of CD3+ and CD19+ cells using magnetic beads (79), and differentiation of functional NKCs from enriched CD34+ progenitors present in cord blood and bone marrow (80, 81). While good manufacturing practice (GMP) guidelines have been established, inconsistencies exist between the cytotoxicity, expansion rate, receptor expression, cytokine secretion and phenotype based on their respective source and expansion method (80, 8284) which may influence their therapeutic activity. iPSCs derived NKCs possess improved expansion rate, cytokine secretion and cytotoxic compared to those from PB. They can also be genetically functionalized to harness tumor targeting, cytolytic activity and persistence in the TME (85). They have been clinically tested for different diseases including graft versus host disease, Parkinson’s disease and heart failure (86, 87). Synergistic activity of iPSCs-derived NKCs with other effector T-cells and successful in vitro tumor infiltration has been described in animal studies (86). Other studies in this regard are focused on developing efficient methods and designing biomaterials for activation, expansion and isolation of NKCs (85, 8891). Gao et al. recently classified the biological and transcriptomic signatures of cord blood and placenta-derived NKCs revealing the cellular/molecular level similarities and differences existing between the NK cell types (92). More of such studies are needed as they serve as database for cell-based immunotherapy and will be beneficial to understanding and categorizing the mechanism of action of different NKCs.

Optimizing persistence and cytolytic activity of natural killer cells

There is a huge need of novel technologies to enhance the activity of NKCs and their interaction with tumors. Consequently, several methods have been proposed. For instance, concomitant use of NK cell adoptive transfer and other therapeutic methods, including T-cells, chemotherapeutic agents, cytokines and immunomodulatory drugs could fortify NKCs against the TME and be synergistic in tumor immunotherapy (9395). The biological targets of these supplementary molecules like cytokines and drugs vary from those of NKCs providing synergy (96) but their safety must also be assured before clinical application. Biber et al. describes the design of a non-viral lipid nanoparticle-based delivery system that encapsulates small interfering RNAs which targets NKCs in vivo, silences inhibitory molecules, and activate NK cell anti-tumor activity (97). Park et al. reports that Aurantii Fructus Immaturus, a commonly used herb in traditional medicine enhances the anticancer efficacy of NK (98), Bispecific killer cells engagers (BiKEs) and trispecific killer cells engagers (TriKEs) improve in vitro secretion of cytokines and efficiently induce the cytotoxic effects of NKCs (99, 100). Moving forwards, optimizing and improving these formulations to avoid undesirable side effects are vital steps toward their clinical application.

Conclusion

The role of NKCs in neutralizing senescent, stressed and malignant cells has attracted enormous research attention aimed at producing clinical grade NKCs for adoptive cell immunotherapy. Currently, some clinical studies are designed to determine the safety and efficacy of ex vivo activated and expanded NKCs while others test the effect of administering the NKCs in combination with other immune molecules. Major advances, including the development of efficient ex vivo expansion systems, prolonged in vivo persistence and genetic manipulation strategies involving CARs are currently explored to facilitate clinically applicable NK cell therapeutics. However, additional research effort is needed to enhance tumor targeting, overcome immune suppression by inhibitory signals or cells and exhaustion in the TME, increase persistence in allogeneic settings, facilitate expansion in patients, sustain in vivo surveillance against tumor relapse, and increase the applicability of NK cell therapy to a wider range of life-threatening diseases especially those marked by depletion in NK cell function. Finally, iPSC-NKCs hold great prospects and further refinement of their differentiation protocol is necessary to match the phenotypic properties of PB NKCs.

Author contributions

Conceptualization, HQ, CY, FY, KT, CX, and ME. Writing—original draft preparation, HQ, ME, RZ, and ST. Writing—review and editing, FY, ME, KT, CX, and ST. Supervision and approval, HQ, CY, and ST. All authors contributed to the article and approved the submitted version.

Conflict of interest

Authors HQ, CY, FY, KT, CX were employed by company NanHua Bio-medicine CO., Ltd.

The remaining authors declare that the research was conducted in the absence of any commercial or financial relationships that could be construed as a potential conflict of interest.

Publisher’s note

All claims expressed in this article are solely those of the authors and do not necessarily represent those of their affiliated organizations, or those of the publisher, the editors and the reviewers. Any product that may be evaluated in this article, or claim that may be made by its manufacturer, is not guaranteed or endorsed by the publisher.

References

1. Hoogduijn MJ, Issa F, Casiraghi F, Reinders MEJ. Cellular therapies in organ transplantation. Transpl Int (2021) 34(2):233–44. doi: 10.1111/tri.13789

PubMed Abstract | CrossRef Full Text | Google Scholar

2. Shang Z, Wang M, Zhang B, Wang X, Wanyan P. Clinical translation of stem cell therapy for spinal cord injury still premature: results from a single-arm meta-analysis based on 62 clinical trials. BMC Med (2022) 20(1):284. doi: 10.1186/s12916-022-02482-2

PubMed Abstract | CrossRef Full Text | Google Scholar

3. Demel I, Koristek Z, Motais B, Hajek R, Jelinek T. Natural killer cells: Innate immune system as a part of adaptive immunotherapy in hematological malignancies. Am J Hematol (2022) 97(6):802–17. doi: 10.1002/ajh.26529

PubMed Abstract | CrossRef Full Text | Google Scholar

4. Cheng M, Chen Y, Xiao W, Sun R, Tian Z. NK cell-based immunotherapy for malignant diseases. Cell Mol Immunol (2013) 10(3):230–52. doi: 10.1038/cmi.2013.10

PubMed Abstract | CrossRef Full Text | Google Scholar

5. Hanna J, Mandelboim O. When killers become helpers. Trends Immunol (2007) 28(5):201–6. doi: 10.1016/j.it.2007.03.005

PubMed Abstract | CrossRef Full Text | Google Scholar

6. Huntington ND, Vosshenrich CA, Di Santo JP. Developmental pathways that generate natural-killer-cell diversity in mice and humans. Nat Rev Immunol (2007) 7(9):703–14. doi: 10.1038/nri2154

PubMed Abstract | CrossRef Full Text | Google Scholar

7. Grégoire C, Chasson L, Luci C, Tomasello E, Geissmann F, Vivier E, et al. The trafficking of natural killer cells. Immunol Rev (2007) 220(1):169–82. doi: 10.1111/j.1600-065X.2007.00563.x

PubMed Abstract | CrossRef Full Text | Google Scholar

8. Damele L, Spaggiari GM, Parodi M, Mingari MC, Vitale M, Vitale C. Cord blood-derived natural killer cell exploitation in immunotherapy protocols: More than a promise? Cancers (Basel) (2022) 14(18):4439–45. doi: 10.3390/cancers14184439

PubMed Abstract | CrossRef Full Text | Google Scholar

9. Sadelain M, Brentjens R, Rivière I. The basic principles of chimeric antigen receptor design. Cancer Discovery (2013) 3(4):388–98. doi: 10.1158/2159-8290.Cd-12-0548

PubMed Abstract | CrossRef Full Text | Google Scholar

10. Goldenson BH, Hor P, Kaufman DS. iPSC-derived natural killer cell therapies - expansion and targeting. Front Immunol (2022) 13:841107. doi: 10.3389/fimmu.2022.841107

PubMed Abstract | CrossRef Full Text | Google Scholar

11. Maddineni S, Silberstein JL, Sunwoo JB. Emerging NK cell therapies for cancer and the promise of next generation engineering of iPSC-derived NK cells. J Immunother Cancer (2022) 10(5):4693–701. doi: 10.1136/jitc-2022-004693

CrossRef Full Text | Google Scholar

12. Jacquelot N, Seillet C, Souza-Fonseca-Guimaraes F, Sacher AG, Belz GT, Ohashi PS. Natural killer cells and type 1 innate lymphoid cells in hepatocellular carcinoma: Current knowledge and future perspectives. Int J Mol Sci (2021) 22(16):9044–50. doi: 10.3390/ijms22169044

PubMed Abstract | CrossRef Full Text | Google Scholar

13. Xue JS, Ding ZN, Meng GX, Yan LJ, Liu H, Li HC, et al. The prognostic value of natural killer cells and their Receptors/Ligands in hepatocellular carcinoma: A systematic review and meta-analysis. Front Immunol (2022) 13:872353. doi: 10.3389/fimmu.2022.872353

PubMed Abstract | CrossRef Full Text | Google Scholar

14. Smyth MJ, Cretney E, Kelly JM, Westwood JA, Street SE, Yagita H, et al. Activation of NK cell cytotoxicity. Mol Immunol (2005) 42(4):501–10. doi: 10.1016/j.molimm.2004.07.034

PubMed Abstract | CrossRef Full Text | Google Scholar

15. Prager I, Liesche C, van Ooijen H, Urlaub D, Verron Q, Sandström N, et al. NK cells switch from granzyme b to death receptor-mediated cytotoxicity during serial killing. J Exp Med (2019) 216(9):2113–27. doi: 10.1084/jem.20181454

PubMed Abstract | CrossRef Full Text | Google Scholar

16. Coënon L, Villalba M. From CD16a biology to antibody-dependent cell-mediated cytotoxicity improvement. Front Immunol (2022) 13:913215. doi: 10.3389/fimmu.2022.913215

PubMed Abstract | CrossRef Full Text | Google Scholar

17. Pende D, Falco M, Vitale M, Cantoni C, Vitale C, Munari E, et al. Killer ig-like receptors (KIRs): Their role in NK cell modulation and developments leading to their clinical exploitation. Front Immunol (2019) 10:1179. doi: 10.3389/fimmu.2019.01179

PubMed Abstract | CrossRef Full Text | Google Scholar

18. Zhao XY, Yu XX, Xu ZL, Cao XH, Huo MR, Zhao XS, et al. Donor and host coexpressing KIR ligands promote NK education after allogeneic hematopoietic stem cell transplantation. Blood Adv (2019) 3(24):4312–25. doi: 10.1182/bloodadvances.2019000242

PubMed Abstract | CrossRef Full Text | Google Scholar

19. Van Acker HH, Capsomidis A, Smits EL, Van Tendeloo VF. CD56 in the immune system: More than a marker for cytotoxicity? Front Immunol (2017) 8:892. doi: 10.3389/fimmu.2017.00892

PubMed Abstract | CrossRef Full Text | Google Scholar

20. Gunesch JT, Dixon AL, Ebrahim TA, Berrien-Elliott MM, Tatineni S, Kumar T, et al. CD56 regulates human NK cell cytotoxicity through Pyk2. Elife (2020) 9:57346–50. doi: 10.7554/eLife.57346

CrossRef Full Text | Google Scholar

21. Schwane V, Huynh-Tran VH, Vollmers S, Yakup VM, Sauter J, Schmidt AH, et al. Distinct signatures in the receptor repertoire discriminate CD56bright and CD56dim natural killer cells. Front Immunol (2020) 11:568927. doi: 10.3389/fimmu.2020.568927

PubMed Abstract | CrossRef Full Text | Google Scholar

22. Michel T, Poli A, Cuapio A, Briquemont B, Iserentant G, Ollert M, et al. Human CD56bright NK cells: An update. J Immunol (2016) 196(7):2923–31. doi: 10.4049/jimmunol.1502570

PubMed Abstract | CrossRef Full Text | Google Scholar

23. Campbell KS, Hasegawa J. Natural killer cell biology: an update and future directions. J Allergy Clin Immunol (2013) 132(3):536–44. doi: 10.1016/j.jaci.2013.07.006

PubMed Abstract | CrossRef Full Text | Google Scholar

24. Morvan MG, Lanier LL. NK cells and cancer: you can teach innate cells new tricks. Nat Rev Cancer (2016) 16(1):7–19. doi: 10.1038/nrc.2015.5

PubMed Abstract | CrossRef Full Text | Google Scholar

25. Beliën J, Goris A, Matthys P. Natural killer cells in multiple sclerosis: Entering the stage. Front Immunol (2022) 13:869447. doi: 10.3389/fimmu.2022.869447

PubMed Abstract | CrossRef Full Text | Google Scholar

26. Soelistyoningsih D, Susianti H, Kalim H, Handono K. The phenotype of CD3-CD56(bright) and CD3-CD56(dim) natural killer cells in systemic lupus erythematosus patients and its relation to disease activity. Reumatologia. (2022) 60(4):258–65. doi: 10.5114/reum.2022.119042

PubMed Abstract | CrossRef Full Text | Google Scholar

27. Mahajan D, Sharma NR, Kancharla S, Kolli P, Tripathy A, Sharma AK, et al. Role of natural killer cells during pregnancy and related complications. Biomolecules (2022) 12(1):68–72. doi: 10.3390/biom12010068

PubMed Abstract | CrossRef Full Text | Google Scholar

28. Halma J, Pierce S, McLennan R, Bradley T, Fischer R. Natural killer cells in liver transplantation: Can we harness the power of the immune checkpoint to promote tolerance? Clin Transl Sci (2022) 15(5):1091–103. doi: 10.1111/cts.13208

PubMed Abstract | CrossRef Full Text | Google Scholar

29. Gebru YA, Gupta H, Kim HS, Eom JA, Kwon GH, Park E, et al. T Cell subsets and natural killer cells in the pathogenesis of nonalcoholic fatty liver disease. Int J Mol Sci (2021) 22(22):12190–5. doi: 10.3390/ijms222212190

PubMed Abstract | CrossRef Full Text | Google Scholar

30. Liu B, Yang GX, Sun Y, Tomiyama T, Zhang W, Leung PSC, et al. Decreased CD57 expression of natural killer cells enhanced cytotoxicity in patients with primary sclerosing cholangitis. Front Immunol (2022) 13:912961. doi: 10.3389/fimmu.2022.912961

PubMed Abstract | CrossRef Full Text | Google Scholar

31. Sun Y, Zhou J, Jiang Y. Negative regulation and protective function of natural killer cells in HIV infection: Two sides of a coin. Front Immunol (2022) 13:842831. doi: 10.3389/fimmu.2022.842831

PubMed Abstract | CrossRef Full Text | Google Scholar

32. Wu X, Xiao Y, Guo D, Zhang Z, Liu M. Reduced NK cell cytotoxicity by papillomatosis-derived TGF-β contributing to low-risk HPV persistence in JORRP patients. Front Immunol (2022) 13:849493. doi: 10.3389/fimmu.2022.849493

PubMed Abstract | CrossRef Full Text | Google Scholar

33. Palano MT, Cucchiara M, Gallazzi M, Riccio F, Mortara L, Gensini GF, et al. When a friend becomes your enemy: Natural killer cells in atherosclerosis and atherosclerosis-associated risk factors. Front Immunol (2021) 12:798155. doi: 10.3389/fimmu.2021.798155

PubMed Abstract | CrossRef Full Text | Google Scholar

34. Meredith PJ, Walford RL. Autoimmunity, histocompatibility, and aging. Mech Ageing Dev (1979) 9(1):61–77. doi: 10.1016/0047-6374(79)90120-9

PubMed Abstract | CrossRef Full Text | Google Scholar

35. Witkowski JM, Larbi A, Le Page A, Fülöp T. Natural killer cells, aging, and vaccination. Interdiscip Top Gerontol Geriatr (2020) 43:18–35. doi: 10.1159/000504493

PubMed Abstract | CrossRef Full Text | Google Scholar

36. Knorr DA, Bachanova V, Verneris MR, Miller JS. Clinical utility of natural killer cells in cancer therapy and transplantation. Semin Immunol (2014) 26(2):161–72. doi: 10.1016/j.smim.2014.02.002

PubMed Abstract | CrossRef Full Text | Google Scholar

37. Du N, Guo F, Wang Y, Cui J. NK cell therapy: A rising star in cancer treatment. Cancers (Basel) (2021) 13(16):4129–34. doi: 10.3390/cancers13164129

PubMed Abstract | CrossRef Full Text | Google Scholar

38. Chan IS, Ewald AJ. The changing role of natural killer cells in cancer metastasis. J Clin Invest (2022) 132(6):143762–7. doi: 10.1172/jci143762

CrossRef Full Text | Google Scholar

39. Lutz CT, Karapetyan A, Al-Attar A, Shelton BJ, Holt KJ, Tucker JH, et al. Human NK cells proliferate and die in vivo more rapidly than T cells in healthy young and elderly adults. J Immunol (2011) 186(8):4590–8. doi: 10.4049/jimmunol.1002732

PubMed Abstract | CrossRef Full Text | Google Scholar

40. Hayhoe RPG, Henson SM, Akbar AN, Palmer DB. Variation of human natural killer cell phenotypes with age: Identification of a unique KLRG1-negative subset. Hum Immunol (2010) 71(7):676–81. doi: 10.1016/j.humimm.2010.03.014

PubMed Abstract | CrossRef Full Text | Google Scholar

41. Remarque EJ, Pawelec G. T-Cell immunosenescence and its clinical relevance in man. Rev Clin Gerontol (1998) 8:5–14.

Google Scholar

42. Kim JS, Choi SE, Yun IH, Kim JY, Ahn C, Kim SJ, et al. Human cytomegalovirus UL18 alleviated human NK-mediated swine endothelial cell lysis. Biochem Biophys Res Commun (2004) 315(1):144–50. doi: 10.1016/j.bbrc.2004.01.027

PubMed Abstract | CrossRef Full Text | Google Scholar

43. Sundström Y, Nilsson C, Lilja G, Kärre K, Troye-Blomberg M, Berg L. The expression of human natural killer cell receptors in early life. Scand J Immunol (2007) 66(2):335–44. doi: 10.1111/j.1365-3083.2007.01980.x

PubMed Abstract | CrossRef Full Text | Google Scholar

44. Béziat V, Nguyen S, Lapusan S, Hervier B, Dhédin N, Bories D, et al. Fully functional NK cells after unrelated cord blood transplantation. Leukemia. (2009) 23:721–8. doi: 10.1038/leu.2008.343

PubMed Abstract | CrossRef Full Text | Google Scholar

45. Sagiv A, Biran A, Yon M, Simon JM, Lowe SW, Krizhanovsky V. Granule exocytosis mediates immune surveillance of senescent cells. Oncogene. (2013) 32:1971–7. doi: 10.1038/onc.2012.206

PubMed Abstract | CrossRef Full Text | Google Scholar

46. Solana R, Alonso MC, Peña J. Natural killer cells in healthy aging. Exp Gerontol (1999) 34(3):435–43. doi: 10.1016/s0531-5565(99)00008-x

PubMed Abstract | CrossRef Full Text | Google Scholar

47. Ravaglia G, Forti P, Maioli F, Bastagli L, Facchini A, Mariani E, et al. Effect of micronutrient status on natural killer cell immune function in healthy free-living subjects aged >/=90 y. Am J Clin Nutr (2000) 71(2):590–8. doi: 10.1093/ajcn/71.2.590

PubMed Abstract | CrossRef Full Text | Google Scholar

48. Ojo-Amaize EA, Conley EJ, Peter JB. Decreased natural killer cell activity is associated with severity of chronic fatigue immune dysfunction syndrome. Clin Infect Dis (1994) 18 Suppl 1:S157–9. doi: 10.1093/clinids/18.supplement_1.s157

PubMed Abstract | CrossRef Full Text | Google Scholar

49. Mocchegiani E, Muzzioli M, Giacconi R, Cipriano C, Gasparini N, Franceschi C, et al. Metallothioneins/PARP-1/IL-6 interplay on natural killer cell activity in elderly: parallelism with nonagenarians and old infected humans. Effect zinc supply Mech Ageing Dev (2003) 124(4):459–68. doi: 10.1016/S0047-6374(03)00023-X

CrossRef Full Text | Google Scholar

50. Kiessling R, Klein E, Pross H, Wigzell H. “Natural” killer cells in the mouse. II. cytotoxic cells with specificity for mouse moloney leukemia cells. characteristics of the killer cell. Eur J Immunol (1975) 5(2):117–21. doi: 10.1002/eji.1830050209

PubMed Abstract | CrossRef Full Text | Google Scholar

51. Weil S, Memmer S, Lechner A, Huppert V, Giannattasio A, Becker T, et al. Natural killer group 2D ligand depletion reconstitutes natural killer cell immunosurveillance of head and neck squamous cell carcinoma. Front Immunol (2017) 8:387. doi: 10.3389/fimmu.2017.00387

PubMed Abstract | CrossRef Full Text | Google Scholar

52. Ramos-Mejia V, Arellano-Galindo J, Mejía-Arangure JM, Cruz-Munoz ME. A NK cell odyssey: From bench to therapeutics against hematological malignancies. Front Immunol (2022) 13:803995. doi: 10.3389/fimmu.2022.803995

PubMed Abstract | CrossRef Full Text | Google Scholar

53. Martín-Antonio B, Suñe G, Perez-Amill L, Castella M, Urbano-Ispizua A. Natural killer cells: Angels and devils for immunotherapy. Int J Mol Sci (2017) 18(9):1868–72. doi: 10.3390/ijms18091868

PubMed Abstract | CrossRef Full Text | Google Scholar

54. Papak I, Chruściel E, Dziubek K, Kurkowiak M, Urban-Wójciuk Z, Marjański T, et al. What inhibits natural killers’ performance in tumour. Int J Mol Sci (2022) 23(13):7030–4. doi: 10.3390/ijms23137030

PubMed Abstract | CrossRef Full Text | Google Scholar

55. Valeri A, García-Ortiz A, Castellano E, Córdoba L, Maroto-Martín E, Encinas J, et al. Overcoming tumor resistance mechanisms in CAR-NK cell therapy. Front Immunol (2022) 13:953849. doi: 10.3389/fimmu.2022.953849

PubMed Abstract | CrossRef Full Text | Google Scholar

56. Ferrari de Andrade L, Tay RE, Pan D, Luoma AM, Ito Y, Badrinath S, et al. Antibody-mediated inhibition of MICA and MICB shedding promotes NK cell-driven tumor immunity. Science. (2018) 359(6383):1537–42. doi: 10.1126/science.aao0505

PubMed Abstract | CrossRef Full Text | Google Scholar

57. Fang F, Xie S, Chen M, Li Y, Yue J, Ma J, et al. Advances in NK cell production. Cell Mol Immunol (2022) 19(4):460–81. doi: 10.1038/s41423-021-00808-3

PubMed Abstract | CrossRef Full Text | Google Scholar

58. Shankar K, Capitini CM, Saha K. Genome engineering of induced pluripotent stem cells to manufacture natural killer cell therapies. Stem Cell Res Ther (2020) 11(1):234. doi: 10.1186/s13287-020-01741-4

PubMed Abstract | CrossRef Full Text | Google Scholar

59. Khawar MB, Sun H. CAR-NK cells: From natural basis to design for kill. Front Immunol (2021) 12:707542. doi: 10.3389/fimmu.2021.707542

PubMed Abstract | CrossRef Full Text | Google Scholar

60. Zhang L, Meng Y, Feng X, Han Z. CAR-NK cells for cancer immunotherapy: from bench to bedside. biomark Res (2022) 10(1):12. doi: 10.1186/s40364-022-00364-6

PubMed Abstract | CrossRef Full Text | Google Scholar

61. Marofi F, Abdul-Rasheed OF, Rahman HS, Budi HS, Jalil AT, Yumashev AV, et al. CAR-NK cell in cancer immunotherapy; a promising frontier. Cancer Sci (2021) 112(9):3427–36. doi: 10.1111/cas.14993

PubMed Abstract | CrossRef Full Text | Google Scholar

62. 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(1):367. doi: 10.1186/s13046-021-02148-6

PubMed Abstract | CrossRef Full Text | Google Scholar

63. Ingegnere T, Mariotti FR, Pelosi A, Quintarelli C, De Angelis B, Tumino N, et al. Human CAR NK cells: A new non-viral method allowing high efficient transfection and strong tumor cell killing. Front Immunol (2019) 10:957. doi: 10.3389/fimmu.2019.00957

PubMed Abstract | CrossRef Full Text | Google Scholar

64. Schmidt P, Raftery MJ, Pecher G. Engineering NK cells for CAR therapy-recent advances in gene transfer methodology. Front Immunol (2020) 11:611163. doi: 10.3389/fimmu.2020.611163

PubMed Abstract | CrossRef Full Text | Google Scholar

65. Kumar A, Lee SJ, Liu Q, Chan AKN, Pokharel SP, Yu J, et al. Generation and validation of CRISPR-engineered human natural killer cell lines for research and therapeutic applications. STAR Protoc (2021) 2(4):100874. doi: 10.1016/j.xpro.2021.100874

PubMed Abstract | CrossRef Full Text | Google Scholar

66. Miller JS, Soignier Y, Panoskaltsis-Mortari A, McNearney SA, Yun GH, Fautsch SK, et al. Successful adoptive transfer and in vivo expansion of human haploidentical NK cells in patients with cancer. Blood. (2005) 105(8):3051–7. doi: 10.1182/blood-2004-07-2974

PubMed Abstract | CrossRef Full Text | Google Scholar

67. Kennedy PR, Felices M, Miller JS. Challenges to the broad application of allogeneic natural killer cell immunotherapy of cancer. Stem Cell Res Ther (2022) 13(1):165. doi: 10.1186/s13287-022-02769-4

PubMed Abstract | CrossRef Full Text | Google Scholar

68. Mylod E, Lysaght J, Conroy MJ. Natural killer cell therapy: A new frontier for obesity-associated cancer. Cancer Letters (2022) 535:215620. doi: 10.1016/j.canlet.2022.215620

PubMed Abstract | CrossRef Full Text | Google Scholar

69. Díaz-Basabe A, Burrello C, Lattanzi G, Botti F, Carrara A, Cassinotti E, et al. Human intestinal and circulating invariant natural killer t cells are cytotoxic against colorectal cancer cells via the perforin-granzyme pathway. Mol Oncol (2021) 15(12):3385–403. doi: 10.1002/1878-0261.13104

PubMed Abstract | CrossRef Full Text | Google Scholar

70. Park JD, Kim KS, Choi SH, Jo GH, Choi JH, Park SW, et al. ELK3 modulates the antitumor efficacy of natural killer cells against triple negative breast cancer by regulating mitochondrial dynamics. J Immunother Cancer (2022) 10(7):4825–35. doi: 10.1136/jitc-2022-004825

CrossRef Full Text | Google Scholar

71. Feinberg D, Ramakrishnan P, Wong DP, Asthana A, Parameswaran R. Inhibition of O-GlcNAcylation decreases the cytotoxic function of natural killer cells. Front Immunol (2022) 13:841299. doi: 10.3389/fimmu.2022.841299

PubMed Abstract | CrossRef Full Text | Google Scholar

72. Eaton-Fitch N, Du Preez S, Cabanas H, Muraki K, Staines D, Marshall-Gradisnik S. Impaired TRPM3-dependent calcium influx and restoration using naltrexone in natural killer cells of myalgic encephalomyelitis/chronic fatigue syndrome patients. J Transl Med (2022) 20(1):94. doi: 10.1186/s12967-022-03297-8

PubMed Abstract | CrossRef Full Text | Google Scholar

73. Medjouel Khlifi H, Guia S, Vivier E, Narni-Mancinelli E. Role of the ITAM-bearing receptors expressed by natural killer cells in cancer. Front Immunol (2022) 13:898745. doi: 10.3389/fimmu.2022.898745

PubMed Abstract | CrossRef Full Text | Google Scholar

74. Wu J, He B, Miao M, Han X, Dai H, Dou H, et al. Enhancing natural killer cell-mediated cancer immunotherapy by the biological macromolecule nocardia rubra cell-wall skeleton. Pathol Oncol Res (2022) 28:1610555. doi: 10.3389/pore.2022.1610555

PubMed Abstract | CrossRef Full Text | Google Scholar

75. Wu Z, Park S, Lau CM, Zhong Y, Sheppard S, Sun JC, et al. Dynamic variability in SHP-1 abundance determines natural killer cell responsiveness. Sci Signal (2021) 14(708):eabe5380. doi: 10.1126/scisignal.abe5380

PubMed Abstract | CrossRef Full Text | Google Scholar

76. Burns LJ, Weisdorf DJ, DeFor TE, Vesole DH, Repka TL, Blazar BR, et al. IL-2-based immunotherapy after autologous transplantation for lymphoma and breast cancer induces immune activation and cytokine release: a phase I/II trial. Bone Marrow Transpl (2003) 32(2):177–86. doi: 10.1038/sj.bmt.1704086

CrossRef Full Text | Google Scholar

77. Holthof LC, Stikvoort A, van der Horst HJ, Gelderloos AT, Poels R, Li F, et al. Bone marrow mesenchymal stromal cell-mediated resistance in multiple myeloma against NK cells can be overcome by introduction of CD38-CAR or TRAIL-variant. Hemasphere. (2021) 5(5):e561. doi: 10.1097/hs9.0000000000000561

PubMed Abstract | CrossRef Full Text | Google Scholar

78. Cho D, Campana D. Expansion and activation of natural killer cells for cancer immunotherapy. Korean J Lab Med (2009) 29(2):89–96. doi: 10.3343/kjlm.2009.29.2.89

PubMed Abstract | CrossRef Full Text | Google Scholar

79. Eissens DN, Schaap NP, Preijers FW, Dolstra H, van Cranenbroek B, Schattenberg AV, et al. CD3+/CD19+-depleted grafts in HLA-matched allogeneic peripheral blood stem cell transplantation lead to early NK cell cytolytic responses and reduced inhibitory activity of NKG2A. Leukemia. (2010) 24(3):583–91. doi: 10.1038/leu.2009.269

PubMed Abstract | CrossRef Full Text | Google Scholar

80. Sarvaria A, Jawdat D, Madrigal JA, Saudemont A. Umbilical cord blood natural killer cells, their characteristics, and potential clinical applications. Front Immunol (2017) 8:329. doi: 10.3389/fimmu.2017.00329

PubMed Abstract | CrossRef Full Text | Google Scholar

81. Yang C, Siebert JR, Burns R, Gerbec ZJ, Bonacci B, Rymaszewski A, et al. Heterogeneity of human bone marrow and blood natural killer cells defined by single-cell transcriptome. Nat Commun (2019) 10(1):3931. doi: 10.1038/s41467-019-11947-7

PubMed Abstract | CrossRef Full Text | Google Scholar

82. Kaur K, Ko MW, Chen F, Jewett A. Defective NK cell expansion, cytotoxicity, and lack of ability to differentiate tumors from a pancreatic cancer patient in a long term follow-up: implication in the progression of cancer. Cancer Immunol Immunother (2022) 71(5):1033–47. doi: 10.1007/s00262-021-03044-w

PubMed Abstract | CrossRef Full Text | Google Scholar

83. Goldenson BH, Zhu H, Wang YM, Heragu N, Bernareggi D, Ruiz-Cisneros A, 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. doi: 10.3389/fimmu.2020.561553

PubMed Abstract | CrossRef Full Text | Google Scholar

84. Luevano M, Daryouzeh M, Alnabhan R, Querol S, Khakoo S, Madrigal A, et al. The unique profile of cord blood natural killer cells balances incomplete maturation and effective killing function upon activation. Hum Immunol (2012) 73(3):248–57. doi: 10.1016/j.humimm.2011.12.015

PubMed Abstract | CrossRef Full Text | Google Scholar

85. Tarannum M, Romee R, Shapiro RM. Innovative strategies to improve the clinical application of NK cell-based immunotherapy. Front Immunol (2022) 13:859177. doi: 10.3389/fimmu.2022.859177

PubMed Abstract | CrossRef Full Text | Google Scholar

86. Karagiannis P, Kim SI. iPSC-derived natural killer cells for cancer immunotherapy. Mol Cells (2021) 44(8):541–8. doi: 10.14348/molcells.2021.0078

PubMed Abstract | CrossRef Full Text | Google Scholar

87. Yamanaka S. Pluripotent stem cell-based cell therapy-promise and challenges. Cell Stem Cell (2020) 27(4):523–31. doi: 10.1016/j.stem.2020.09.014

PubMed Abstract | CrossRef Full Text | Google Scholar

88. Lin C, Zhang J. Reformation in chimeric antigen receptor based cancer immunotherapy: Redirecting natural killer cell. Biochim Biophys Acta Rev Cancer (2018) 1869(2):200–15. doi: 10.1016/j.bbcan.2018.01.005

PubMed Abstract | CrossRef Full Text | Google Scholar

89. Berrien-Elliott MM, Romee R, Fehniger TA. Improving natural killer cell cancer immunotherapy. Curr Opin Organ Transpl (2015) 20(6):671–80. doi: 10.1097/mot.0000000000000243

CrossRef Full Text | Google Scholar

90. 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 Biochem Biophys (2015) 73(3):723–9. doi: 10.1007/s12013-015-0688-3

PubMed Abstract | CrossRef Full Text | Google Scholar

91. Kweon S, Phan MT, Chun S, Yu H, Kim J, Kim S, et al. Expansion of human NK cells using K562 cells expressing OX40 ligand and short exposure to IL-21. Front Immunol (2019) 10:879. doi: 10.3389/fimmu.2019.00879

PubMed Abstract | CrossRef Full Text | Google Scholar

92. Gao H, Liu M, Zhang Y, Zhang L, Xie B. Multifaceted characterization of the biological and transcriptomic signatures of natural killer cells derived from cord blood and placental blood. Cancer Cell Int (2022) 22(1):291. doi: 10.1186/s12935-022-02697-6

PubMed Abstract | CrossRef Full Text | Google Scholar

93. Cifaldi L, Locatelli F, Marasco E, Moretta L, Pistoia V. Boosting natural killer cell-based immunotherapy with anticancer drugs: a perspective. Trends Mol Med (2017) 23(12):1156–75. doi: 10.1016/j.molmed.2017.10.002

PubMed Abstract | CrossRef Full Text | Google Scholar

94. Floros T, Tarhini AA. Anticancer cytokines: Biology and clinical effects of interferon-α2, interleukin (IL)-2, IL-15, IL-21, and IL-12. Semin Oncol (2015) 42(4):539–48. doi: 10.1053/j.seminoncol.2015.05.015

PubMed Abstract | CrossRef Full Text | Google Scholar

95. Ishikawa T, Okayama T, Sakamoto N, Ideno M, Oka K, Enoki T, et al. Phase I clinical trial of adoptive transfer of expanded natural killer cells in combination with IgG1 antibody in patients with gastric or colorectal cancer. Int J Cancer (2018) 142(12):2599–609. doi: 10.1002/ijc.31285

PubMed Abstract | CrossRef Full Text | Google Scholar

96. Nielsen CM, Wolf A-S, Goodier MR, Riley EM. Synergy between common γ chain family cytokines and IL-18 potentiates innate and adaptive pathways of NK cell activation. Front Immunol (2016) 7:101. doi: 10.3389/fimmu.2016.00101

PubMed Abstract | CrossRef Full Text | Google Scholar

97. Biber G, Sabag B, Raiff A, Ben-Shmuel A, Puthenveetil A, Benichou JIC, et al. Modulation of intrinsic inhibitory checkpoints using nano-carriers to unleash NK cell activity. EMBO Mol Med (2022) 14(1):e14073. doi: 10.15252/emmm.202114073

PubMed Abstract | CrossRef Full Text | Google Scholar

98. Park A, Yang Y, Lee Y, Jung H, Kim TD, Noh JY, et al. Aurantii fructus immaturus enhances natural killer cytolytic activity and anticancer efficacy in vitro and in vivo. Front Med (Lausanne) (2022) 9:973681. doi: 10.3389/fmed.2022.973681

PubMed Abstract | CrossRef Full Text | Google Scholar

99. Demaria O, Gauthier L, Debroas G, Vivier E. Natural killer cell engagers in cancer immunotherapy: Next generation of immuno-oncology treatments. Eur J Immunol (2021) 51(8):1934–42. doi: 10.1002/eji.202048953

PubMed Abstract | CrossRef Full Text | Google Scholar

100. Wiernik A, Foley B, Zhang B, Verneris MR, Warlick E, Gleason MK, et al. Targeting natural killer cells to acute myeloid leukemia in vitro with a CD16 x 33 bispecific killer cell engager and ADAM17 inhibition. Clin Cancer Res (2013) 19(14):3844–55. doi: 10.1158/1078-0432.Ccr-13-0505

PubMed Abstract | CrossRef Full Text | Google Scholar

Keywords: natural killer (NK) cells, immunosenescence, immunotherapy, cancer, chemotherapy, biotherapeutics

Citation: Qin H, You C, Yan F, Tan K, Xu C, Zhao R, Ekpo MD and Tan S (2022) Overcoming the challenges in translational development of natural killer cell therapeutics: An opinion paper. Front. Oncol. 12:1062765. doi: 10.3389/fonc.2022.1062765

Received: 06 October 2022; Accepted: 22 November 2022;
Published: 02 December 2022.

Edited by:

Ye Li, University of Texas MD Anderson Cancer Center, United States

Reviewed by:

Sjoukje Van Der Stegen, Memorial Sloan Kettering Cancer Center, United States

Copyright © 2022 Qin, You, Yan, Tan, Xu, Zhao, Ekpo and Tan. This is an open-access article distributed under the terms of the Creative Commons Attribution License (CC BY). The use, distribution or reproduction in other forums is permitted, provided the original author(s) and the copyright owner(s) are credited and that the original publication in this journal is cited, in accordance with accepted academic practice. No use, distribution or reproduction is permitted which does not comply with these terms.

*Correspondence: Changqiao You, youcq@landfar.cn; Marlene Davis Ekpo, marlenedavis@csu.edu.cn

Disclaimer: All claims expressed in this article are solely those of the authors and do not necessarily represent those of their affiliated organizations, or those of the publisher, the editors and the reviewers. Any product that may be evaluated in this article or claim that may be made by its manufacturer is not guaranteed or endorsed by the publisher.