Open access peer-reviewed chapter

Natural Killer Cells for Cancer Immunotherapy: Opportunities and Challenges

Written By

Leisheng Zhang, Xiaoming Feng, Zhihai Han and Zhongchao Han

Submitted: 20 September 2022 Reviewed: 12 December 2022 Published: 29 December 2022

DOI: 10.5772/intechopen.109480

From the Edited Volume

Natural Killer Cells - Lessons and Challenges

Edited by Leisheng Zhang

Chapter metrics overview

119 Chapter Downloads

View Full Metrics

Abstract

Natural killer (NK) cells are advantaged immune cells and play a pivotal role in both innate and adaptive immune responses. To date, autogenous and allogenic NK cells have been generated from a variety of origins, including perinatal blood (e.g., umbilical cord blood and placental blood), peripheral blood, and even stem cells (hematopoietic stem cells and pluripotent stem cells). NK cells function mainly via antibody-dependent cell-mediated cytotoxicity (ADCC), direct cytolytic effect, and paracrine effects (e.g., IFN-γ, GM-CSF, granzyme, and perforin). Distinguishing from the adaptive immunizing cells (e.g., T and B lymphocytes), NK cells, and chimeric antigen receptor-transduced NK (CAR-NK), cell-based cytotherapy is adequate to fulfill the biofunction of eliminating pathogenic infection, combating hematological malignancies and metastatic solid tumors, and delaying aging. In this chapter, we mainly focus on the state-of-the-art renewal of NK cell-based cytotherapy for cancer immunosurveillance and immunotherapy from the view of high-efficient in vitro preparation (e.g., candidate cell sources and ex vivo cultivation) and preclinical and clinical investigation. Furthermore, we also figure out the promising prospects and the concomitant challenges of NK cell-based remedies for cancer management in future, which will collectively benefit the development of NK cell-based cancer immunotherapy in future.

Keywords

  • natural killer cells
  • cancer immunotherapy
  • CAR-NK cells
  • preclinical and clinical investigation

1. Introduction

Cancer, including hematological malignancies and metastatic solid tumors, is one of the leading causes of death worldwide with increasing incidence and mortality, which has thus become a major public health issue and heavy burden to patients and their families [1, 2]. For example, a total of over 4,568,000 cases were newly diagnosed cancer in China, and over 3,002,000 cancer deaths occurred in 2020 [3]. Of them, lung cancer is the most frequent type, followed by colorectal cancer and gastric cancer, whereas lung cancer, hepatocellular carcinoma, and stomach cancers are the top three most deadly subtypes of cancers in the general population.

For decades, depicting the cancer pattern has also been acknowledged to provide basic know-hows for developing novel strategies for cancer management [4, 5]. To date, a variety of modifiable risk factors for cancer have been identified to fuel the shift of cancer transition, such as oncogenes, air pollution, smoking, alcohol consumption, unhealthful dietary habits, obesity, physical inactivity, infectious agents, diabetes, improved livelihood, and even rapid economic development [6, 7, 8]. For example, cancer-related fatigue is one of the most distressing and prevalent symptoms among patients, which usually results in great research challenges [9].

For decades, effective implementation strategies have been developed to optimize the advancements in the fields of tumor survivorship, diagnosis, treatment, and end-of-life care [10]. For instance, a series of treatments have been developed to conquer cancers with high uncontrolled cell division and heterogeneity, including surgery (e.g., laparoscopic rectal surgery and robotic surgery) [11, 12], photothermal therapy (PTT) [13, 14, 15], radiotherapy [16], chemotherapy [17], oncolytic virotherapy [18], RNA vaccine [19, 20], hormone therapy [21], peptide-based neoantigen vaccine [22, 23], gene therapy [24, 25, 26], immunotherapy (e.g., immune cells and checkpoint inhibitors) [2], and even nanomaterial-mediated nanotheranostics (e.g., organic nanomaterials, inorganic nanomaterials, and organic–inorganic hybrid nanomaterials). Nevertheless, the aforementioned strategies have also revealed inherent shortcomings (e.g., severe toxicity, off-target effects, graft-versus-host disease, and drug delivery barriers), which collectively hinder the further improvement in cancer administration [27, 28].

State-of-the-art renewal has highlighted the feasibility of cellular immunotherapy as promising remedy for cancer administration on the basis of the rapid progress of cryobiology and oncobiology [29, 30, 31, 32, 33]. For instance, pioneering and talented investigators in the field have conducted systematic and detailed studies from bench to bedside to conquer hematologic tumors and solid tumors, including the non-gene-edited natural killer (NK) cells and the CAR-transduced counterparts (e.g., CAR-T, CAR-NK cells). Distinguishing from the adaptive T cells, NK cells are adequate to efficaciously fulfill the requirement of combating the aforementioned metastatic solid tumors and hematological malignancies via antibody-dependent cell-mediated cytotoxicity (ADCC), direct cytolytic effect, and paracrine effects dispense with antigen presentation [2, 28, 33, 34].

Therewith, the overview of this literature aimed to summarize the current knowledge and scope of the evidence on NK cell-based immunotherapy for cancer patients from the view of cell source (e.g., peripheral blood, perinatal blood, and cell lines), ex vivo amplification and activation (e.g., coculture and monolayer culture), mode of action (e.g., ADCC and cytolytic effect), together with preclinical and clinical practice of NK cell-based cancer immunotherapy. Meanwhile, we also indicate the promising prospects and the concomitant challenges of NK cell-based cancer immunotherapy in regenerative medicine.

Advertisement

2. Natural killer (NK) cells

NK cells are pivotal part of the innate immune defense against cancer and infection, and in particular, in combating certain bacterial and viral pathogens [35]. In the 1970s, Kiessling and their colleagues firstly identified NK cells from a mouse with advantaged characteristics of cytolytic effector dispense with preliminary antigen presentation [36, 37]. Incipiently, NK cells were considered critical components to play an important role in host innate immunity and the first line of defense, and in particular, in the antiviral infection [38, 39]. To date, NK cells have been recognized as the third type of lymphocyte with vital roles in both innate and adaptive immune responses to malignancies and viral infection, which also function in delaying aging and benefiting hematopoietic reconstitution [28, 40].

As a heterogeneous lymphocyte subpopulation, NK cells can be divided into the cytotoxic CD3CD56dimCD16high subset and the IFN-γ-producing CD3CD56brightCD16+ counterpart [28, 41]. Of note, state-of-the-art updates have also put forward the existence of long-lived memory-like NK cells with reinforced responsiveness against cancer and the CD16-dependent functional capability [42, 43].

2.1 Cell sources for NK cell generation

To date, NK cells have been generated from various origins, including umbilical cord blood (UCB) [44], placental blood [44], peripheral blood (PB)-derived mononuclear cells (PBMCs) [43, 45], NK cell lines (e.g., NK-92, YT) [28], and even derived from CD34+ hematopoietic stem cells (HSCs) [28], human embryonic stem cells (hESCs), and induced pluripotent stem cells (iPSCs) [34, 46]. Interestingly, the content and cytotoxic activity of NK cells in perinatal and peripheral blood vary a lot. For example, NK cells take a proportion of PB cells ranging from 5–20% [47, 48], whereas the proportions of NK cells in UCB and placental blood were less than 5% [49] and 2% [50], respectively. Meanwhile, over 60% of NK cells in UCB and PB reveal the CD3 CD56+ CD16+ subset, whereas the rest belongs to the CD3 CD56+ CD16 subset. Conversely, only 40% of NK cells in placental blood are CD3 CD56+ CD16+ cells compared with the rest CD3CD56+CD16 subset with a percentage of 60% instead [50]. Additionally, Dogra and the colleagues found that immune responses of NK cells in diverse tissue sites were different, which indicated the tissue localization in the regulation of the NK cell development as well as the biofunctions involved in antiviral and tumor immunosurveillance [51].

During the past decades, NK cells were isolated and identified from PB to determine the specific characteristics associated with anti-infection and anticancer as well as pathogenesis (e.g., recurrent miscarriage and hematologic malignancies) [45]. PB-derived NK (PB-NK) cells have been recognized as cytotoxic innate lymphocytes, which are endowed with a unique capacity to kill a broad spectrum of virus-infected cells, cancers, and even chronic obstructive pulmonary disease (COPD) [52, 53]. Currently, PBMCs are considered the dominant ingredient for clinical- or GMP- grade NK cell manufacturing [48]. However, the stability and yield of NK cell production are far from adequate largely attributed to the limitations in blood donators and the present procedures [43, 54]. Distinguishing from those of PB-NK cells, perinatal blood-derived NK cells reveal advantaged properties such as vigorous cytolytic activity, preferable cellular vitality, and enhanced cytotoxicity [28]. Of note, the latest kinds of literature have further indicated the large-scale preparation of NK cells from human pluripotent stem cells (hPSCs), which are considered excellent candidates for “off-the-shelf” anticancer immunotherapy and NK cell development [55].

2.2 Ex vivo preparation of NK cells

Strategies for ex vivo expansion of NK cells have allowed preparing enough amounts of NK cells for clinical trials and new drug application (NDA). However, current donor cell-dependent strategies for ex vivo preparation of NK cells can only produce limited amount of “made-to-order” therapeutic cells for limited patients [46, 56]. For decades, a series of methods have been developed for large-scale ex vivo NK cell preparation, including cell sorting, feeder cell coculture (e.g., K562 cells), gene editing, low oxygen treatment, multiple cytokine cocktail stimulation, three-dimensional rotation, and bioreactors [28]. For instance, Mu et al. developed a simple, safe, and cost-effective strategy for ex vivo purification and expansion of NK cells from CB via zoledronate and streptococcus stimulation, and dispense of cell sorting or feeder cells/multiple cytokines within 21 days [57]. Alves et al. reported the ex vivo expansion of CD56+ lymphocytes, including NK cells and NKT-like subset, from PBMCs in CellGro medium supplemented with IL-2 and fetal bovine serum (FBS), anti-CD3 (9–10 initial days) for 21 days [56]. Meanwhile, distinguishing from the resting subset, the expanded NK cells showed a higher expression level of activating receptors (e.g., CD16, NKp44, NKp46, NKG2D, NKp30, CD62L, and CD69) and a higher concentration of cytokine secretion (e.g., IFN-γ, TNF-α, and GM-CSF). Instead, Radice and the colleagues verified that PB-derived NK cells from inpatients with early-stage colorectal cancer showed enhanced ex vivo cytotoxicity against cancer cell lines (e.g., HT-29, K562, and human CRC Caco-2) after the stimulation of low-doses of sequential-kinetic-activated (SKA) interferon-γ (IFN-γ) in vitro [58].

Interestingly, we took advantage of the “3ILs”-based cell programming and induced high-efficient generation of NK cells from peripheral and perinatal blood within 14 days [43, 44]. Of note, we and other investigators in the field found that the expanded NK cells revealed elevated content and enhanced cytotoxicity against various tumor cell lines in vitro compared to the resident counterpart [44, 57]. Additionally, Klingemann and the colleagues tested the feasibility of a broadly applicable method for blood progenitor cell and NK cell preparation based on the well-established NK-92 cell line, which is derived from a patient with non-Hodgkin’s lymphoma [59]. Collectively, NK cell expansion and activation require multiple cell signal cascades for proliferation, survival, and activation. Therewith, most of the current ex vivo expansion strategies are mainly focused on either using genetically modified allogeneic feeder cells or substituting the indicated factors by utilizing the autologous feeder cells [60].

2.3 Mode of action for NK cell-based cytotherapy

Aforesaid, autologous and allogeneic NK cells act as critical effector lymphocytes and mediate cancer immune clearance surveillance [61]. Generally, NK cells spontaneously function in abnormal cell elimination, pathogenic microorganism, and tumor immunosurveillance via ADCC, direct cytolytic effect, paracrine of cytokines, and receptor-ligand-associated natural cytotoxicity effects as well [48, 62, 63].

Generally, the mode of action for NK cell-based cytotherapy is mainly based on “NK cell education,” which has also been recognized as functional maturation of NK cells. As recently reviewed by Zhang and the colleagues, this process usually involves a series of step-wise intermediate stages and obtains self-major histocompatibility complex class I (MHC-I) recognition [64]. Meanwhile, as mentioned above, the IFN-γ-producing CD3CD56bright NK cells were less mature when compared with the cytotoxic CD3CD56dim subset [41]. Interestingly, new insights into the long-lived memory-like NK cells indicated the enhanced CD16-dependent functional capability as well as responsiveness, which thus hold advantages over the CD25- subset in NK cell-based interventions against cancer [42, 43].

Advertisement

3. Chimeric antigen receptor-transduced NK (CAR-NK) cells

In recent years, CAR-transduced NK (CAR-NK) cells are considered as novel therapeutic options to reduce the incidence of relapse and recurrent cancers. A number of preclinical and clinical investigations have indicated CAR-NK cells as “off-the-shelf” products for cancer immunotherapy attributed to CAR-dependent and NK cell receptor-dependent signaling pathways [34]. Differing from those of CAR-T cells, CAR-NK cells avoid the adverse effect during cancer administration such as immune cell-associated neurotoxicity syndrome (ICANS), acute cytokine release syndrome (CRS), and graft-versus-host disease (GVHD), which thus represent the therapeutic paradigm for boosting the immune system to reinforce anticancer responses and eventually eliminate malignancies [34].

To date, a considerable number of cancer-specific antigens and the concomitant costimulatory molecules as well as the well-established subsets (e.g., NKp44, NKp46, and NKG2D) have been involved in CAR-NK cell-based immunotherapy. For example, the existing CARs transduced to generate CAR-T cells were directly transferred to NK cells, including 4-1BB, CD19, CD20, CD22, CD276, BCMA, CD3ζ, and CD28 [28, 65, 66, 67, 68, 69, 70]. Simultaneously, pioneering and talented investigators in the field have dedicated themselves to optimize the constructs of CARs to enhance the efficacy of anticancer effects [32, 71]. For example, suicide genes have been integrated into the fourth generation of CARs to remove the unanticipated toxicity [72]. Furthermore, due to the advances in genome-editing techniques, the applicability and feasibility of CAR-NK cells have vastly accelerated cancer immunotherapy. Despite with deficiency in CAR constructs delivery into NK cells, a plethora of groups have highlighted the possibility for high-efficient CAR transduction via Lentivirus [28, 73, 74], retrovirus [75, 76], and even nonviral-mediated transfection (e.g., lipofection and electroporation, PiggyBac, and the sleeping beauty (SB) subsets) [77, 78]. Collectively, recent developments in the clinical practice of genetically modified NK cells with chimeric antigen receptors (CARs) are promising and challenging for NK cell-based immunotherapy.

Advertisement

4. NK cell-based cancer immunotherapy

Cancer immunotherapy with diverse preclinical and clinical attempts has been attractive and received considerable attention [79]. As time passes by, the feasibility of using NK cells in the treatment of human disorders such as refractory lung diseases, pathogenic infection, hematopoietic reconstitution, hematological malignancies, and metastatic solid tumors has increased in recent years [80]. In particular, the antileukemic potential of NK cells has raised considerable interest among researchers and clinicians for hematological malignancy administration. Nowadays, immune-based remedies characterized by the infusion of ex vivo activated or freshly isolated NK cells have become a research hotspot in cancer immunotherapy [34, 81, 82, 83].

In the 1980s, the dysfunction of NK cells was reported with a higher incidence of cancers, including Chédiak–Higashi syndrome and X-linked lymphoproliferative syndrome, which was first indicated in cancer immunosurveillance [41, 84, 85]. Recently, Bryce et al. found that NK cells in patients with chronic lymphocytic leukemia (CLL) showed reduction in content and abnormality in gene expression pattern (e.g., GATA-1, GATA-2, PU.1, and HIF-1α) in bone marrow [86].

To date, nearly 900 NK cell-based clinical trials, including the observational and interventional subtypes, were registered upon multiple tumors according to the Clinicaltrials.gov website. Generally, most of the trials were involved in hematologic malignancy administration, including acute myeloid leukemia (AML), acute lymphoblastic leukemia (ALL), chronic myelogenous leukemia (CML), and myelodysplastic syndromes (MDS). Meanwhile, clinical pieces of evidence have also indicated the therapeutic effect of NK cell-based cytotherapy upon various solid tumors such as gastric carcinoma, colorectal cancer, liver cancer, and squamous cell lung cancer [87, 88, 89]. However, the definite NK cell-based immunotherapy upon metastatic solid tumors has not been verified, which was largely attributed to the dysregulation of NK cell function and immune subversion by various factors (e.g., IDO, PGE2, and TGF-β) in the tumor microenvironment [48].

Advertisement

5. Discussion and conclusions

Nowadays, with the rapid progress of available remedies for cancer treatments, patients are adequate to live with the consequences of cancer. NK cells have been regarded as the first line of innate immune cells, which are involved in providing the surveillance and elimination of the stressed, infected, damaged, and malignant cells [90, 91]. NK cell-based cancer immunotherapy has largely benefited the treatments of patients for longer living and better outcomes, which also supplies new preferences for cancer administration and helps improve decisions about optimal treatment. Of note, recent kinds of literature have suggested the promising aspect of achieving considerable amount of NK cells for adoptive immunotherapy after a period of ex vivo expansion with multifaceted stimulation. Nevertheless, the improvement of ex vivo expansion procedures for the clinical application of NK cells generated under good manufacturing practice conditions is of prime importance in future.

Advertisement

Acknowledgments

The coauthors thank the members of Key Laboratory of Molecular Diagnostics and Precision Medicine for Surgical Oncology in Gansu Province and NHC Key Laboratory of Diagnosis and Therapy of Gastrointestinal Tumor, Gansu Provincial Hospital, Hefei Institute of Physical Science in Chinese Academy of Sciences, The First Affiliated Hospital of Shandong First Medical University, Health-Biotech (Tianjin) Stem Cell Research Institute Co., Ltd., and Jiangxi Health-Biotech Stem Cell Technology Co., Ltd. for their technical support. This work was supported by grants from the National Natural Science Foundation of China (No. 82260031), the project Youth Fund supported by Shandong Provincial Natural Science Foundation (ZR2020QC097), Fujian Provincial Ministerial Finance Special Project (2021XH018), Science and technology projects of Guizhou Province (QKH-J-ZK[2021]-107), The 2021 Central-Guided Local Science and Technology Development Fund (ZYYDDFFZZJ-1), Medical Innovation Project of Fujian Provincial Health and Health Commission (2019-CX-21), Jiangxi Provincial Natural Science Foundation (20224BAB206077, 20212BAB216073), Key project funded by Department of Science and Technology of Shangrao City (2020AB002, 2020 K003, 2021F013), Jiangxi Provincial Key New Product Incubation Program Funded by Technical Innovation Guidance Program of Shangrao City (2020G002), the Non-profit Central Research Institute Fund of Chinese Academy of Medical Sciences (2019PT320005), and Horizontal Project upon Retrospective Analyses of COVID-19 (2020XH001).

Advertisement

Conflict of interest

The authors declare no conflict of interest.

Advertisement

Notes/thanks/other declarations

Not applicable.

Advertisement

Abbreviation

ADCCAntibody-dependent cell-mediated cytotoxicity
CAR-NKChimeric antigen receptor-transduced NK
NKNatural killer
PTTPhotothermal therapy
CAR-TCAR-transduced T cells
UCBUmbilical cord blood
PBPeripheral blood
PBMCsPeripheral blood-derived mononuclear cells
PB-NKPeripheral blood-derived NK
COPDChronic obstructive pulmonary disease
NDANew drug application
FBSFetal bovine serum
HSCsHematopoietic stem cells
CRSCytokine release syndrome
GVHDGraft-versus-host disease
ICANSImmune cell-associated neurotoxicity syndrome
GMPGood manufacturing practices
CMLChronic myelogenous leukemia
hPSCsHuman pluripotent stem cells
MDSMyelodysplastic syndromes
ALLAcute lymphoblastic leukemia
hESCsHuman embryonic stem cells
hiPSCsHuman induced PSCs
AMLAcute myeloid leukemia
CLLChronic lymphocytic leukemia
SKASequential-kinetic-activated
IFN-γInterferon-γ

References

  1. 1. Chen W, Zheng R, Baade PD, Zhang S, Zeng H, Bray F, et al. Cancer statistics in China, 2015. CA: a Cancer Journal for Clinicians. 2016;66(2):115-132
  2. 2. Cany J, van der Waart AB, Tordoir M, Franssen GM, Hangalapura BN, de Vries J, et al. Natural killer cells generated from cord blood hematopoietic progenitor cells efficiently target bone marrow-residing human leukemia cells in NOD/SCID/IL2Rg(null) mice. PLoS One. 2013;8(6):e64384
  3. 3. Qiu H, Cao S, Xu R. Cancer incidence, mortality, and burden in China: A time-trend analysis and comparison with the United States and United Kingdom based on the global epidemiological data released in. Cancer Communication, (Lond). 2020;41(10):1037-1048
  4. 4. Feng RM, Zong YN, Cao SM, Xu RH. Current cancer situation in China: Good or bad news from the 2018 global cancer statistics? Cancer Communication, (Lond). 2019;39(1):22
  5. 5. Wei W, Zeng H, Zheng R, Zhang S, An L, Chen R, et al. Cancer registration in China and its role in cancer prevention and control. The Lancet Oncology. 2020;21(7):e342-e349
  6. 6. Sun D, Li H, Cao M, He S, Lei L, Peng J, et al. Cancer burden in China: Trends, risk factors and prevention. Cancer Biology & Medicine. 2020;17(4):879-895
  7. 7. Yin P, Brauer M, Cohen AJ, Wang H, Li J, Burnett RT, et al. The effect of air pollution on deaths, disease burden, and life expectancy across China and its provinces, 1990-2017: An analysis for the global burden of disease study 2017. Lancet Planet Health. 2020;4(9):e386-e398
  8. 8. Maomao C, He L, Dianqin S, Siyi H, Xinxin Y, Fan Y, et al. Current cancer burden in China: Epidemiology, etiology, and prevention. Cancer Biology & Medicine. 2022;19(8):1121-1138
  9. 9. Belloni S, Arrigoni C, Caruso R. Effects from physical exercise on reduced cancer-related fatigue: A systematic review of systematic reviews and meta-analysis. Acta Oncologica. 2021;60(12):1678-1687
  10. 10. Brouwers MC, Garcia K, Makarski J, Daraz L, Evidence Expert P. Team KTfCCiCPR: The landscape of knowledge translation interventions in cancer control: What do we know and where to next? A review of systematic reviews. Implementation Science. 2011;6:130
  11. 11. Rowcroft A, Joh D, Pandanaboyana S, Loveday B. Hepato-pancreato-biliary and transplant surgery experience among New Zealand general surgery trainees. ANZ Journal of Surgery. 2021;91(6):1125-1130
  12. 12. Ramshaw BJ. Laparoscopic surgery for cancer patients. CA: a Cancer Journal for Clinicians. 1997;47(6):327-350
  13. 13. Li X, Lovell JF, Yoon J, Chen X. Clinical development and potential of photothermal and photodynamic therapies for cancer. Nature Reviews. Clinical Oncology. 2020;17(11):657-674
  14. 14. Cheng L, Zhang X, Tang J, Lv Q , Liu J. Gene-engineered exosomes-thermosensitive liposomes hybrid nanovesicles by the blockade of CD47 signal for combined photothermal therapy and cancer immunotherapy. Biomaterials. 2021;275:120964
  15. 15. Kadkhoda J, Tarighatnia A, Tohidkia MR, Nader ND, Aghanejad A. Photothermal therapy-mediated autophagy in breast cancer treatment: Progress and trends. Life Sciences. 2022;298:120499
  16. 16. Olivares-Urbano MA, Grinan-Lison C, Marchal JA, Nunez MI. CSC Radioresistance: A therapeutic challenge to improve radiotherapy effectiveness in cancer. Cell. 2020;9(7):1651
  17. 17. Schirrmacher V. From chemotherapy to biological therapy: A review of novel concepts to reduce the side effects of systemic cancer treatment (review). International Journal of Oncology. 2019;54(2):407-419
  18. 18. Kwan A, Winder N, Muthana M. Oncolytic Virotherapy treatment of breast cancer: Barriers and recent advances. Viruses. 2021;13(6):1128
  19. 19. Miao L, Zhang Y, Huang L. mRNA vaccine for cancer immunotherapy. Molecular Cancer. 2021;20(1):41
  20. 20. Sahin U, Oehm P, Derhovanessian E, Jabulowsky RA, Vormehr M, Gold M, et al. An RNA vaccine drives immunity in checkpoint-inhibitor-treated melanoma. Nature. 2020;585(7823):107-112
  21. 21. Deli T, Orosz M, Jakab A. Hormone replacement therapy in cancer survivors - review of the literature. Pathology Oncology Research. 2020;26(1):63-78
  22. 22. Ding Z, Li Q , Zhang R, Xie L, Shu Y, Gao S, et al. Personalized neoantigen pulsed dendritic cell vaccine for advanced lung cancer. Signal Transduction and Targeted Therapy. 2021;6(1):26
  23. 23. Jou J, Harrington KJ, Zocca MB, Ehrnrooth E, Cohen EEW. The changing landscape of therapeutic cancer vaccines-novel platforms and Neoantigen identification. Clinical Cancer Research. 2021;27(3):689-703
  24. 24. Cross D, Burmester JK. Gene therapy for cancer treatment: Past, present and future. Clinical Medicine & Research. 2006;4(3):218-227
  25. 25. Graham TA, Sottoriva A. Measuring cancer evolution from the genome. The Journal of Pathology. 2017;241(2):183-191
  26. 26. Lee YT, Tan YJ, Oon CE. Molecular targeted therapy: Treating cancer with specificity. European Journal of Pharmacology. 2018;834:188-196
  27. 27. Mun EJ, Babiker HM, Weinberg U, Kirson ED, Von Hoff DD. Tumor-treating fields: A fourth modality in cancer treatment. Clinical Cancer Research. 2018;24(2):266-275
  28. 28. Zhang L, Liu M, Yang S, Wang J, Feng X, Han Z. Natural killer cells: Of-the-shelf cytotherapy for cancer immunosurveillance. American Journal of Cancer Research. 2021;11(4):1770-1791
  29. 29. Depil S, Duchateau P, Grupp SA, Mufti G, Poirot L. 'Off-the-shelf' allogeneic CAR T cells: Development and challenges. Nature Reviews. Drug Discovery. 2020;19(3):185-199
  30. 30. Hernandez-Lopez A, Tellez-Gonzalez MA, Mondragon-Teran P, Meneses-Acosta A. Chimeric antigen receptor-T cells: A pharmaceutical scope. Frontiers in Pharmacology. 2021;12:720692
  31. 31. Liu E, Marin D, Banerjee P, Macapinlac HA, Thompson P, Basar R, et al. Use of CAR-transduced natural killer cells in CD19-positive lymphoid tumors. The New England Journal of Medicine. 2020;382(6):545-553
  32. 32. Siegler EL, Zhu Y, Wang P, Yang L. Off-the-shelf CAR-NK cells for cancer immunotherapy. Cell Stem Cell. 2018;23(2):160-161
  33. 33. Wrona E, Borowiec M, Potemski P. CAR-NK cells in the treatment of solid tumors. International Journal of Molecular Sciences. 2021;22(11):5899
  34. 34. Zhang L, Meng Y, Feng X, Han Z. CAR-NK cells for cancer immunotherapy: From bench to bedside. Biomarker Research. 2022;10(1):12
  35. 35. Orange JS. Natural killer cell deficiency. The Journal of Allergy and Clinical Immunology. 2013;132(3):515-525
  36. 36. Kiessling R, Klein E, Wigzell H. "natural" killer cells in the mouse. I. Cytotoxic cells with specificity for mouse Moloney leukemia cells. Specificity and distribution according to genotype. European Journal of Immunology. 1975;5(2):112-117
  37. 37. 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. European Journal of Immunology. 1975;5(2):117-121
  38. 38. Wu F, Xie M, Hun M, She Z, Li C, Luo S, et al. Natural killer cell-derived extracellular vesicles: Novel players in cancer immunotherapy. Frontiers in Immunology. 2021;12:658698
  39. 39. Mancini M, Vidal SM. Mechanisms of natural killer cell evasion through viral adaptation. Annual Review of Immunology. 2020;38:511-539
  40. 40. O'Brien KL, Finlay DK. Immunometabolism and natural killer cell responses. Nature Reviews. Immunology. 2019;19(5):282-290
  41. 41. Morvan MG, Lanier LL. NK cells and cancer: You can teach innate cells new tricks. Nature Reviews. Cancer. 2016;16(1):7-19
  42. 42. Capuano C, Battella S, Pighi C, Franchitti L, Turriziani O, Morrone S, et al. Tumor-targeting anti-CD20 antibodies mediate In vitro expansion of memory natural killer cells: Impact of CD16 affinity ligation conditions and In vivo priming. Frontiers in Immunology. 2018;9:1031
  43. 43. Liu M, Meng Y, Zhang L, Han Z, Feng X. High-efficient generation of natural killer cells from peripheral blood with preferable cell vitality and enhanced cytotoxicity by combination of IL-2, IL-15 and IL-18. Biochemical and Biophysical Research Communications. 2021;534:149-156
  44. 44. Zhang L, Liu M, Song B, Miao W, Zhan R, Yang S, et al. Decoding the multidimensional signatures of resident and expanded natural killer cells generated from perinatal blood. American Journal of Cancer Research. 2022;12(5):2132-2145
  45. 45. Guerrero B, Hassouneh F, Delgado E, Casado JG, Tarazona R. Natural killer cells in recurrent miscarriage: An overview. Journal of Reproductive Immunology. 2020;142:103209
  46. 46. Zeng J, Tang SY, Toh LL, Wang S. Generation of "off-the-shelf" natural killer cells from peripheral blood cell-derived induced pluripotent stem cells. Stem Cell Reports. 2017;9(6):1796-1812
  47. 47. Koepsell SA, Miller JS, McKenna DH Jr. Natural killer cells: A review of manufacturing and clinical utility. Transfusion. 2013;53(2):404-410
  48. 48. Guillerey C, Huntington ND, Smyth MJ. Targeting natural killer cells in cancer immunotherapy. Nature Immunology. 2016;17(9):1025-1036
  49. 49. Balassa K, Rocha V. Anticancer cellular immunotherapies derived from umbilical cord blood. Expert Opinion on Biological Therapy. 2018;18(2):121-134
  50. 50. Kang L, Voskinarian-Berse V, Law E, Reddin T, Bhatia M, Hariri A, et al. Characterization and ex vivo expansion of human placenta-derived natural killer cells for cancer immunotherapy. Frontiers in Immunology. 2013;4:101
  51. 51. Dogra P, Rancan C, Ma W, Toth M, Senda T, Carpenter DJ, et al. Tissue determinants of human NK cell development, function, and residence. Cell. 2020;180(4):749-763 e713
  52. 52. Kim JM, Yi E, Cho H, Choi WS, Ko DH, Yoon DH, et al. Assessment of NK cell activity based on NK cell-specific receptor synergy in peripheral blood mononuclear cells and whole blood. International Journal of Molecular Sciences. 2020;21(21):8112
  53. 53. Rao Y, Le Y, Xiong J, Pei Y, Sun Y. NK cells in the pathogenesis of chronic obstructive pulmonary disease. Frontiers in Immunology. 2021;12:666045
  54. 54. Blum KS, Pabst R. Lymphocyte numbers and subsets in the human blood. Do they mirror the situation in all organs? Immunology Letters. 2007;108(1):45-51
  55. 55. Li Y, Hermanson DL, Moriarity BS, Kaufman DS. Human iPSC-derived natural killer cells engineered with chimeric antigen receptors enhance anti-tumor activity. Cell Stem Cell. 2018;23(2):181-192 e185
  56. 56. Alves PCM, De Angelo Andrade LAL, Petta CA, Lorand-Metze I, Derchain SF, Guimaraes F. Ex vivo expansion of CD56+ NK and NKT-like lymphocytes from peripheral blood mononuclear cells of patients with ovarian neoplasia. Scandinavian Journal of Immunology. 2011;74(3):244-252
  57. 57. Mu YX, Zhao YX, Li BY, Bao HJ, Jiang H, Qi XL, et al. A simple method for in vitro preparation of natural killer cells from cord blood. BMC Biotechnology. 2019;19(1):80
  58. 58. Radice E, Miranda V, Bellone G. Low-doses of sequential-kinetic-activated interferon-gamma enhance the ex vivo cytotoxicity of peripheral blood natural killer cells from patients with early-stage colorectal cancer. A preliminary study. Int Immunopharmacol. 2014;19(1):66-73
  59. 59. Klingemann HG, Wong E, Maki G. A cytotoxic NK-cell line (NK-92) for ex vivo purging of leukemia from blood. Biology of Blood and Marrow Transplantation. 1996;2(2):68-75
  60. 60. Becker PS, Suck G, Nowakowska P, Ullrich E, Seifried E, Bader P, et al. Selection and expansion of natural killer cells for NK cell-based immunotherapy. Cancer Immunology, Immunotherapy. 2016;65(4):477-484
  61. 61. Woan KV, Miller JS. Harnessing natural killer cell antitumor immunity: From the bench to bedside. Cancer Immunology Research. 2019;7(11):1742-1747
  62. 62. Freud AG, Mundy-Bosse BL, Yu J, Caligiuri MA. The broad Spectrum of human natural killer cell diversity. Immunity. 2017;47(5):820-833
  63. 63. Hodgins JJ, Khan ST, Park MM, Auer RC, Ardolino M. Killers 2.0: NK cell therapies at the forefront of cancer control. The Journal of Clinical Investigation. 2019;129(9):3499-3510
  64. 64. Vivier E. What is natural in natural killer cells? Immunology Letters. 2006;107(1):1-7
  65. 65. Jayaraman J, Mellody MP, Hou AJ, Desai RP, Fung AW, Pham AHT, et al. CAR-T design: Elements and their synergistic function. eBioMedicine. 2020;58:102931
  66. 66. Zhang Y, Li Y, Cao W, Wang F, Xie X, Li Y, et al. Single-cell analysis of target antigens of CAR-T reveals a potential landscape of "on-target, off-tumor toxicity". Frontiers in Immunology. 2021;12:799206
  67. 67. Genssler S, Burger MC, Zhang C, Oelsner S, Mildenberger I, Wagner M, et al. Dual targeting of glioblastoma with chimeric antigen receptor-engineered natural killer cells overcomes heterogeneity of target antigen expression and enhances antitumor activity and survival. Oncoimmunology. 2016;5(4):e1119354
  68. 68. Imai C, Iwamoto S, Campana D. Genetic modification of primary natural killer cells overcomes inhibitory signals and induces specific killing of leukemic cells. Blood. 2005;106(1):376-383
  69. 69. Xu Y, Liu Q , Zhong M, Wang Z, Chen Z, Zhang Y, et al. 2B4 costimulatory domain enhancing cytotoxic ability of anti-CD5 chimeric antigen receptor engineered natural killer cells against T cell malignancies. Journal of Hematology & Oncology. 2019;12(1):49
  70. 70. Romanski A, Uherek C, Bug G, Seifried E, Klingemann H, Wels WS, et al. CD19-CAR engineered NK-92 cells are sufficient to overcome NK cell resistance in B-cell malignancies. Journal of Cellular and Molecular Medicine. 2016;20(7):1287-1294
  71. 71. Sanmamed MF, Chen L. A paradigm shift in cancer immunotherapy: From enhancement to normalization. Cell. 2018;175(2):313-326
  72. 72. Daher M, Rezvani K. Outlook for new CAR-based therapies with a focus on CAR NK cells: What lies beyond CAR-engineered T cells in the race against cancer. Cancer Discovery. 2021;11(1):45-58
  73. 73. Oelsner S, Friede ME, Zhang C, Wagner J, Badura S, Bader P, et al. Continuously expanding CAR NK-92 cells display selective cytotoxicity against B-cell leukemia and lymphoma. Cytotherapy. 2017;19(2):235-249
  74. 74. Boissel L, Betancur-Boissel M, Lu W, Krause DS, Van Etten RA, Wels WS, et al. Retargeting NK-92 cells by means of CD19- and CD20-specific chimeric antigen receptors compares favorably with antibody-dependent cellular cytotoxicity. Oncoimmunology. 2013;2(10):e26527
  75. 75. Imamura M, Shook D, Kamiya T, Shimasaki N, Chai SM, Coustan-Smith E, et al. Autonomous growth and increased cytotoxicity of natural killer cells expressing membrane-bound interleukin-15. Blood. 2014;124(7):1081-1088
  76. 76. Guven H, Konstantinidis KV, Alici E, Aints A, Abedi-Valugerdi M, Christensson B, et al. Efficient gene transfer into primary human natural killer cells by retroviral transduction. Experimental Hematology. 2005;33(11):1320-1328
  77. 77. Rostovskaya M, Fu J, Obst M, Baer I, Weidlich S, Wang H, et al. Transposon-mediated BAC transgenesis in human ES cells. Nucleic Acids Research. 2012;40(19):e150
  78. 78. Vargas JE, Chicaybam L, Stein RT, Tanuri A, Delgado-Canedo A, Bonamino MH. Retroviral vectors and transposons for stable gene therapy: Advances, current challenges and perspectives. Journal of Translational Medicine. 2016;14(1):288
  79. 79. Yang YJ, Park JC, Kim HK, Kang JH, Park SY. A trial of autologous ex vivo-expanded NK cell-enriched lymphocytes with docetaxel in patients with advanced non-small cell lung cancer as second- or third-line treatment: Phase IIa study. Anticancer Research. 2013;33(5):2115-2122
  80. 80. Guimaraes F, Guven H, Donati D, Christensson B, Ljunggren HG, Bejarano MT, et al. Evaluation of ex vivo expanded human NK cells on antileukemia activity in SCID-beige mice. Leukemia. 2006;20(5):833-839
  81. 81. Yu SJ, Ma C, Heinrich B, Brown ZJ, Sandhu M, Zhang Q , et al. Targeting the crosstalk between cytokine-induced killer cells and myeloid-derived suppressor cells in hepatocellular carcinoma. Journal of Hepatology. 2019;70(3):449-457
  82. 82. Zhang C, Burger MC, Jennewein L, Genßler S, Schönfeld K, Zeiner P, et al. ErbB2/HER2-specific NK cells for targeted therapy of glioblastoma. Journal of the National Cancer Institute. 6 Dec 2015;108(5)
  83. 83. Zhang Q , Zhang H, Ding J, Liu H, Li H, Li H, et al. Combination therapy with EpCAM-CAR-NK-92 cells and Regorafenib against human colorectal cancer models. Journal of Immunology Research. 2018;2018:4263520
  84. 84. Roder JC, Haliotis T, Klein M, Korec S, Jett JR, Ortaldo J, et al. A new immunodeficiency disorder in humans involving NK cells. Nature. 1980;284(5756):553-555
  85. 85. Sullivan JL, Byron KS, Brewster FE, Purtilo DT. Deficient natural killer cell activity in x-linked lymphoproliferative syndrome. Science. 1980;210(4469):543-545
  86. 86. Manso BA, Zhang H, Mikkelson MG, Gwin KA, Secreto CR, Ding W, et al. Bone marrow hematopoietic dysfunction in untreated chronic lymphocytic leukemia patients. Leukemia. 2019;33(3):638-652
  87. 87. Ishigami S, Natsugoe S, Tokuda K, Nakajo A, Xiangming C, Iwashige H, et al. Clinical impact of intratumoral natural killer cell and dendritic cell infiltration in gastric cancer. Cancer Letters. 2000;159(1):103-108
  88. 88. Villegas FR, Coca S, Villarrubia VG, Jimenez R, Chillon MJ, Jareno J, et al. Prognostic significance of tumor infiltrating natural killer cells subset CD57 in patients with squamous cell lung cancer. Lung Cancer. 2002;35(1):23-28
  89. 89. Zheng X, Qian Y, Fu B, Jiao D, Jiang Y, Chen P, et al. Mitochondrial fragmentation limits NK cell-based tumor immunosurveillance. Nature Immunology. 2019;20(12):1656-1667
  90. 90. Huang RS, Lai MC, Lin S. Ex vivo expansion and CRISPR-Cas9 genome editing of primary human natural killer cells. Current Protocols. 2021;1(9):e246
  91. 91. Heipertz EL, Zynda ER, Stav-Noraas TE, Hungler AD, Boucher SE, Kaur N, et al. Current perspectives on "off-the-shelf" allogeneic NK and CAR-NK cell therapies. Frontiers in Immunology. 2021;12:732135

Written By

Leisheng Zhang, Xiaoming Feng, Zhihai Han and Zhongchao Han

Submitted: 20 September 2022 Reviewed: 12 December 2022 Published: 29 December 2022