We use cookies to improve your experience. By continuing to browse this site, you accept our cookie policy.×
Skip main navigation
Aging Health
Bioelectronics in Medicine
Biomarkers in Medicine
Breast Cancer Management
CNS Oncology
Colorectal Cancer
Concussion
Epigenomics
Future Cardiology
Future Medicine AI
Future Microbiology
Future Neurology
Future Oncology
Future Rare Diseases
Future Virology
Hepatic Oncology
HIV Therapy
Immunotherapy
International Journal of Endocrine Oncology
International Journal of Hematologic Oncology
Journal of 3D Printing in Medicine
Lung Cancer Management
Melanoma Management
Nanomedicine
Neurodegenerative Disease Management
Pain Management
Pediatric Health
Personalized Medicine
Pharmacogenomics
Regenerative Medicine

Cancer immunotherapy via stem cell-derived NK cells

    Fatemeh Hosseini

    Department of Applied Cell Sciences, School of Advanced Technologies in Medicine, Tehran University of Medical Sciences, Tehran, 1417755469, Iran

    Research Institute for Oncology, Hematology & Cell Therapy, Shariati Hospital, Tehran University of Medical Sciences, 1416634793, Iran

    ,
    Mohammad Ahmadvand

    Cell Therapy & Hematopoietic Stem Cell Transplantation Research Center, Tehran University of Medical Sciences, Tehran, 1416634793, Iran

    Research Institute for Oncology, Hematology & Cell Therapy, Tehran University of Medical Sciences, Tehran, 1411713135, Iran

    ,
    Roya Karimi

    Department of Tissue Engineering & Applied Cell Sciences, School of Advanced Technologies in Medicine, Tehran University of Medical Sciences, Tehran, 1417755469, Iran

    ,
    Seyed Asadollah Mousavi

    Research Institute for Oncology, Hematology & Cell Therapy, Shariati Hospital, Tehran University of Medical Sciences, 1416634793, Iran

    ,
    Jafar Ai

    *Author for correspondence:

    E-mail Address: jafar_ai@tums.ac.ir

    Department of Applied Cell Sciences, School of Advanced Technologies in Medicine, Tehran University of Medical Sciences, Tehran, 1417755469, Iran

    &
    Mohsen Nikbakht

    **Author for correspondence:

    E-mail Address: m-nikbakht@sina.tums.ac.ir

    Research Institute for Oncology, Hematology & Cell Therapy, Shariati Hospital, Tehran University of Medical Sciences, 1416634793, Iran

    Department of Tissue Engineering & Applied Cell Sciences, School of Advanced Technologies in Medicine, Tehran University of Medical Sciences, Tehran, 1417755469, Iran

    Published Online:https://doi.org/10.2217/imt-2022-0224

    NK cells are the first sentinels of the immune system that can recognize and eradicate transformed cells. Their activation without a need for additional signaling have attracted great attention on the use of NK cells as a promising option in cancer immunotherapy. However, the large-scale production of NK cells for successful NK cells therapy is a challenge that needs to be tackled. Engineering NK cells to avoid tumor escape and improve their antitumor potency are the other matters of focus that have widely been studied in the recent years. This paper reviews the most recent advances in the stem cell-derived NK cell technology and discusses the potential of the engineered NK cells for clinical applications in cancer immunotherapy.

    Plain language summary

    NK cells are important cells in our body's defense system that can find and destroy tumor cells. These cells are made in bone marrow (in adults) or umbilical cord (in the embryonic period) from a population of cells called stem cells, and then released into the blood and lymph. Stem cells are the early ancestral cells that can differentiate into multiple cell types. Because the number and function of NK cells in a tumor context are reduced, thus we can use these stem cells to make lots of NK cells for treatment purposes. Scientists can also make these cells even better at killing tumors by changing them to have special sensors. In the end, NK cells are like superheroes that fight and kill tumor cells, and using stem cells to make them is a really promising way to help treat malignant diseases.

    Papers of special note have been highlighted as: • of interest; •• of considerable interest

    References

    • 1. Wolf NK, Kissiov DU, Raulet DH. Roles of natural killer cells in immunity to cancer, and applications to immunotherapy. Nat. Rev. Immunol. 23(2), 90–105 (2023).
    • 2. Sainiteesh M, John LS, John BS. Emerging NK cell therapies for cancer and the promise of next generation engineering of iPSC-derived NK cells. J. Immunother. Cancer 10(5), e004693 (2022).
    • 3. Shimasaki N, Jain A, Campana D. NK cells for cancer immunotherapy. Nat. Rev. Drug Discov. 19(3), 200–218 (2020).
    • 4. Wu SY, Fu T, Jiang YZ, Shao ZM. Natural killer cells in cancer biology and therapy. Mol. Cancer 19(1), 120 (2020).
    • 5. Chiossone L, Dumas P-Y, Vienne M, Vivier E. Natural killer cells and other innate lymphoid cells in cancer. Nat. Rev. Immunol. 18(11), 671–688 (2018).
    • 6. Rezvani K. Adoptive cell therapy using engineered natural killer cells. Bone Marrow Transplant. 54(2), 785–788 (2019). • A timely review in the field.
    • 7. Rubnitz JE, Inaba H, Ribeiro RC et al. NKAML: a pilot study to determine the safety and feasibility of haploidentical natural killer cell transplantation in childhood acute myeloid leukemia. J. Clin. Oncol. 28(6), 955 (2010). • A pioneer study in the field.
    • 8. Alkhaled M, Elazzazy A, Almaghrabia O. In vivo developing immunotherapy of ex vivo expansion of human natural killer cells from thalassemia patients for leukaemia. Indian J. Pharm. Sci. 84(2), 42–47 (2022).
    • 9. Guillerey C, Huntington ND, Smyth MJ. Targeting natural killer cells in cancer immunotherapy. Nat. Immunol. 17(9), 1025–1036 (2016).
    • 10. Salagianni M, Baxevanis CN, Papamichail M, Perez SA. New insights into the role of NK cells in cancer immunotherapy. Oncoimmunology 1(2), 205–207 (2012).
    • 11. Lian G, Mak TS-K, Yu X, Lan H-Y. Challenges and recent advances in NK cell-targeted immunotherapies in solid tumors. Int. J. Mol. Sci. 23(1), 164 (2021).
    • 12. 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 23(2), 181–192.e5 (2018).
    • 13. Zhu H, Kaufman DS. Engineered human pluripotent stem cell-derived natural killer cells: the next frontier for cancer immunotherapy. Blood Sci. 1(1), 4–11 (2019).
    • 14. Morvan MG, Lanier LL. NK cells and cancer: you can teach innate cells new tricks. Nat. Rev. Cancer 16(1), 7–19 (2016).
    • 15. Crinier A, Narni-Mancinelli E, Ugolini S, Vivier E. SnapShot: natural killer cells. Cell 180(6), 1280–1280.e1 (2020).
    • 16. Jacobs R, Hintzen G, Kemper A et al. CD56bright cells differ in their KIR repertoire and cytotoxic features from CD56dim NK cells. Eur. J. Immunol. 31(10), 3121–3126 (2001).
    • 17. Raulet DH, Vance RE, McMahon CW. Regulation of the natural killer cell receptor repertoire. Annu. Rev. Immunol. 19, 291 (2001).
    • 18. Long EO, Kim HS, Liu D, Peterson ME, Rajagopalan S. Controlling NK cell responses: integration of signals for activation and inhibition. Annu. Rev. Immunol. 31, 227–258 (2013).
    • 19. Ljunggren H-G, Kärre K. In search of the ‘missing self’: MHC molecules and NK cell recognition. Immunol. Today 11, 237–244 (1990).
    • 20. Martínez-Lostao L, Anel A, Pardo J. How do cytotoxic lymphocytes kill cancer cells? Clin. Cancer Res. 21(22), 5047–5056 (2015).
    • 21. Vujanovic NL. Role of TNF family ligands in antitumor activity of natural killer cells. Int. Rev. Immunol. 20(3–4), 415–437 (2001).
    • 22. Müller L, Aigner P, Stoiber D. Type I interferons and natural killer cell regulation in cancer. Front. Immunol. 8, 304 (2017).
    • 23. Yeap WH, Wong KL, Shimasaki N et al. CD16 is indispensable for antibody-dependent cellular cytotoxicity by human monocytes. Sci. Rep. 6(1), 1–22 (2016).
    • 24. Shin MH, Kim J, Lim SA, Kim J, Kim S-J, Lee K-M. NK cell-based immunotherapies in cancer. Immune Netw. 20(2), e14 (2020).
    • 25. Hegde UP, Mukherji B. Current status of chimeric antigen receptor engineered T cell-based and immune checkpoint blockade-based cancer immunotherapies. Cancer Immunol. Immunother. 66(9), 1113–1121 (2017).
    • 26. Hartmann J, Schüßler-Lenz M, Bondanza A, Buchholz CJ. Clinical development of CAR T cells – challenges and opportunities in translating innovative treatment concepts. EMBO Mol. Med. 9(9), 1183–1197 (2017).
    • 27. June C, Lim WA. The principles of engineering immune cells to treat cancer. Cell 168(4), 724–740 (2017).
    • 28. June CH, O'Connor RS, Kawalekar OU, Ghassemi S, Milone MC. CAR T cell immunotherapy for human cancer. Science 359(6382), 1361–1365 (2018).
    • 29. Yang Y, Jacoby E, Fry TJ. Challenges and opportunities of allogeneic donor-derived CAR T cells. Curr. Opin. Hematol. 22(6), 509 (2015).
    • 30. Acharya UH, Dhawale T, Yun S et al. Management of cytokine release syndrome and neurotoxicity in chimeric antigen receptor (CAR) T cell therapy. Expert Rev. Hematol. 12(3), 195–205 (2019).
    • 31. Miller JS, Soignier Y, Panoskaltsis-Mortari A et al. Successful adoptive transfer and in vivo expansion of human haploidentical NK cells in patients with cancer. Blood 105(8), 3051–3057 (2005). • A pioneer study in the field.
    • 32. Du W, Cui L, Zhang J et al. Generation of universal natural killer cells from a cryopreserved cord blood mononuclear cell-derived induced pluripotent stem cell library. FEBS Open Bio 12(10), 1771–1781 (2022).
    • 33. Della Chiesa M, Falco M, Podesta M et al. Phenotypic and functional heterogeneity of human NK cells developing after umbilical cord blood transplantation: a role for human cytomegalovirus? Blood 119(2), 399–410 (2012). • A study that guides the field.
    • 34. Kimbrel EA, Lu S-J. Potential clinical applications for human pluripotent stem cell-derived blood components. Stem Cells Int. 2011, doi: 10.4061/2011/273076 (2011) (Online).
    • 35. Sarvaria A, Jawdat D, Madrigal JA, Saudemont A. Umbilical cord blood natural killer cells, their characteristics, and potential clinical applications. Front. Immunol. 8, 329 (2017).
    • 36. Karagiannis P, Kim S-I. iPSC-derived natural killer cells for cancer immunotherapy. Mol. Cells 44(8), 541 (2021).
    • 37. Zhu H, Kaufman DS. Engineered human pluripotent stem cell-derived natural killer cells: the next frontier for cancer immunotherapy. Blood Sci. 1(01), 4–11 (2019).
    • 38. Knorr DA, Ni Z, Hermanson D et al. Clinical-scale derivation of natural killer cells from human pluripotent stem cells for cancer therapy. Stem Cells Transl. Med. 2(4), 274–283 (2013).
    • 39. Zhang L, Liu M, Yang S, Wang J, Feng X, Han Z. Natural killer cells: of-the-shelf cytotherapy for cancer immunosurveillance. Am. J. Cancer Res. 11(4), 1770–1791 (2021).
    • 40. Uppendahl LD, Felices M, Bendzick L et al. Cytokine-induced memory-like natural killer cells have enhanced function, proliferation, and in vivo expansion against ovarian cancer cells. Gynecol. Oncol. 153(1), 149–157 (2019).
    • 41. Ferrara JL, Levine JE, Reddy P, Holler E. Graft-versus-host disease. Lancet 373(9674), 1550–1561 (2009).
    • 42. Siegler U, Meyer-Monard S, Jörger S et al. Good manufacturing practice-compliant cell sorting and large-scale expansion of single KIR-positive alloreactive human natural killer cells for multiple infusions to leukemia patients. Cytotherapy 12(6), 750–763 (2010).
    • 43. Hermanson DL, Ni Z, Kaufman DS. Human pluripotent stem cells as a renewable source of natural killer cells. In: Hematopoietic Differentiation of Human Pluripotent Stem Cells. Springer, NY, USA, 69–79 (2015).
    • 44. Hsu L-J, Liu C-L, Kuo M-L, Shen C-N, Shen C-R. An alternative cell therapy for cancers: induced pluripotent stem cell (iPSC)-derived natural killer cells. Biomedicines 9(10), 1323 (2021).
    • 45. Woll PS, Grzywacz B, Tian X et al. Human embryonic stem cells differentiate into a homogeneous population of natural killer cells with potent in vivo antitumor activity. Blood 113(24), 6094–6101 (2009).
    • 46. 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 23(2), 181–192.e5 (2018). •• A pioneer study on the use of chimeric antigen receptor-expressing induced pluripotent stem cells-derived NK cells.
    • 47. Alireza P. Evaluation of anti-cancer function in natural killer cells generated from rat hematopoietic stem cells. Thesis for PhD fulfilment, KULA LUMPUR, Malaya. Universiti Malaya. (2020).
    • 48. Armstrong R. Spin Embryoid Bodies as an Improved Method of Blood Cell Differentiation in Human Embryonic Stem Cells and Induced Pluripotent Stem Cells. University of Minnesota, MN, USA (2010).
    • 49. Denman CJ, Senyukov VV, Somanchi SS et al. Membrane-bound IL-21 promotes sustained ex vivo proliferation of human natural killer cells. PlOS ONE 7(1), e30264 (2012).
    • 50. Oberoi P, Kamenjarin K, Villena Ossa JF, Uherek B, Bönig H, Wels WS. Directed differentiation of mobilized hematopoietic stem and progenitor cells into functional NK cells with enhanced antitumor activity. Cells 9(4), 811 (2020).
    • 51. Hermanson DL, Bendzick L, Pribyl L, McCullar V, Vogel RI, Miller JS et al. Induced pluripotent stem cell-derived natural killer cells for treatment of ovarian cancer. Stem Cells 34(1), 93–101 (2016). •• Potential of induced pluripotent stem cells-derived NK cells in cancer treatment.
    • 52. Shankar K, Capitini CM, Saha K. Genome engineering of induced pluripotent stem cells to manufacture natural killer cell therapies. Stem Cell Res. Ther. 11(1), 1–14 (2020).
    • 53. Ni Z, Knorr DA, Bendzick L, Allred J, Kaufman DS. Expression of chimeric receptor CD4 ζ by natural killer cells derived from human pluripotent stem cells improves in vitro activity but does not enhance suppression of HIV infection in vivo. Stem Cells 32(4), 1021–1031 (2014).
    • 54. Abreu TR, Fonseca NA, Gonçalves N, Moreira JN. Current challenges and emerging opportunities of CAR-T cell therapies. J. Control. Rel. 319, 246–261 (2020).
    • 55. Wang L, Dou M, Ma Q, Yao R, Liu J. Chimeric antigen receptor (CAR)-modified NK cells against cancer: opportunities and challenges. Int. Immunopharmacol. 74, doi: 10.1016/j.intimp.2019.105695 (2019) (Online).
    • 56. Ruggeri L, Capanni M, Casucci M et al. Role of natural killer cell alloreactivity in HLA-mismatched hematopoietic stem cell transplantation. Blood 94(1), 333–339 (1999).
    • 57. Ruggeri L, Capanni M, Urbani E et al. Effectiveness of donor natural killer cell alloreactivity in mismatched hematopoietic transplants. Science 295(5562), 2097–2100 (2002).
    • 58. Rezvani K, Rouce R, Liu E, Shpall E. Engineering natural killer cells for cancer immunotherapy. Mol. Ther. 25(8), 1769–1781 (2017).
    • 59. Imai C, Iwamoto S, Campana D. Genetic modification of primary natural killer cells overcomes inhibitory signals and induces specific killing of leukemic cells. Blood 106(1), 376–383 (2005).
    • 60. Galat Y, Elcheva I, Dambaeva S et al. Application of small molecule CHIR99021 leads to the loss of hemangioblast progenitor and increased hematopoiesis of human pluripotent stem cells. Exp. Hematol. 65, 38–48.e1 (2018).
    • 61. Mangal JL, Handlos JL, Esrafili A et al. Engineering metabolism of chimeric antigen receptor (CAR) cells for developing efficient immunotherapies. Cancers 13(5), 1123 (2021).
    • 62. Wang J, Lupo KB, Chambers AM, Matosevic S. Purinergic targeting enhances immunotherapy of CD73+ solid tumors with piggyBac-engineered chimeric antigen receptor natural killer cells. J. Immunother. Cancer 6(1), 1–14 (2018).
    • 63. 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 Res. 73(6), 1777–1786 (2013).
    • 64. Shimasaki N, Fujisaki H, Cho D et al. A clinically adaptable method to enhance the cytotoxicity of natural killer cells against B-cell malignancies. Cytotherapy 14(7), 830–840 (2012).
    • 65. Liu E, Tong Y, Dotti G et al. Cord blood NK cells engineered to express IL-15 and a CD19-targeted CAR show long-term persistence and potent antitumor activity. Leukemia 32(2), 520–531 (2018).
    • 66. Han J, Chu J, Keung Chan W et al. CAR-engineered NK cells targeting wild-type EGFR and EGFRvIII enhance killing of glioblastoma and patient-derived glioblastoma stem cells. Sci. Rep. 5(1), 1–13 (2015).
    • 67. Xu Y, Liu Q, Zhong M et al. 2B4 costimulatory domain enhancing cytotoxic ability of anti-CD5 chimeric antigen receptor engineered natural killer cells against T cell malignancies. J. Hematol. Oncol. 12(1), 1–13 (2019).
    • 68. Schnalzger TE, de Groot MH, Zhang C et al. 3D model for CAR-mediated cytotoxicity using patient-derived colorectal cancer organoids. EMBO J. 38(12), e100928 (2019).
    • 69. Schönfeld K, Sahm C, Zhang C et al. Selective inhibition of tumor growth by clonal NK cells expressing an ErbB2/HER2-specific chimeric antigen receptor. Mol. Ther. 23(2), 330–338 (2015).
    • 70. Biederstädt A, Rezvani K. Engineering the next generation of CAR-NK immunotherapies. Int. J. Hematol. 114(5), 554–571 (2021).
    • 71. Yong CS, John LB, Devaud C et al. A role for multiple chimeric antigen receptor-expressing leukocytes in antigen-specific responses to cancer. Oncotarget 7(23), 34582–34598 (2016).
    • 72. Zhang C, Hu Y, Xiao W, Tian Z. Chimeric antigen receptor-and natural killer cell receptor-engineered innate killer cells in cancer immunotherapy. Cell. Mol. Immunol. 18(9), 2083–2100 (2021).
    • 73. Berraondo P, Sanmamed MF, Ochoa MC et al. Cytokines in clinical cancer immunotherapy. Br. J. Cancer 120(1), 6–15 (2019).
    • 74. Lotze MT, Grimm EA, Mazumder A, Strausser JL, Rosenberg SA. Lysis of fresh and cultured autologous tumor by human lymphocytes cultured in T-cell growth factor. Cancer Res. 41(11 Part 1), 4420–4425 (1981).
    • 75. McMichael EL, Jaime-Ramirez AC, Guenterberg KD et al. IL-21 enhances natural killer cell response to cetuximab-coated pancreatic tumor cells. Clin. Cancer Res. 23(2), 489–502 (2017).
    • 76. Li Q, Ye L-J, Ren H-L et al. Multiple effects of IL-21 on human NK cells in ex vivo expansion. Immunobiology 220(7), 876–888 (2015).
    • 77. Carson WE, Giri JG, Lindemann MJ et al. Interleukin (IL) 15 is a novel cytokine that activates human natural killer cells via components of the IL-2 receptor. J. Exp. Med. 180(4), 1395–1403 (1994).
    • 78. Heinze A, Grebe B, Bremm M et al. The synergistic use of IL-15 and IL-21 for the generation of NK cells from CD3/CD19-depleted grafts improves their ex vivo expansion and cytotoxic potential against neuroblastoma: perspective for optimized immunotherapy post haploidentical stem cell transplantation. Front. Immunol. 10, 2816 (2019).
    • 79. Reina Ortiz C, Bernal AA. Expansion of NK Cells for the Treatment of Multiple Myeloma. University of Zaragoza, CIEN, Zaragoza, Spain, MSc. Thesis (2018).
    • 80. Choi YH, Lim EJ, Kim SW, Moon YW, Park KS, An H-J. IL-27 enhances IL-15/IL-18-mediated activation of human natural killer cells. J. Immunother. Cancer 7(1), 1–12 (2019).
    • 81. Dolstra H, Roeven MW, Spanholtz J et al. Successful transfer of umbilical cord blood CD34+ hematopoietic stem and progenitor-derived NK cells in older acute myeloid leukemia patients. Clin. Cancer Res. 23(15), 4107–4118 (2017).
    • 82. Handgretinger R, Lang P, André MC. Exploitation of natural killer cells for the treatment of acute leukemia. Blood 127(26), 3341–3349 (2016).
    • 83. Zhu H, Lai Y-S, Li Y, Blum RH, Kaufman DS. Concise review: human pluripotent stem cells to produce cell-based cancer immunotherapy. Stem Cells 36(2), 134–145 (2018).
    • 84. Williams BA, Law AD, Routy B et al. A phase I trial of NK-92 cells for refractory hematological malignancies relapsing after autologous hematopoietic cell transplantation shows safety and evidence of efficacy. Oncotarget 8(51), 89256–89268 (2017).
    • 85. Geller MA, Cooley S, Judson PL et al. A phase II study of allogeneic natural killer cell therapy to treat patients with recurrent ovarian and breast cancer. Cytotherapy 13(1), 98–107 (2011).
    • 86. Chambers AM, Lupo KB, Matosevic S. Tumor microenvironment-induced immunometabolic reprogramming of natural killer cells. Front. Immunol. 9, 2517 (2018).
    • 87. Hosseini R, Sarvnaz H, Arabpour M et al. Cancer exosomes and natural killer cells dysfunction: biological roles, clinical significance and implications for immunotherapy. Mol. Cancer 21(1), 15 (2022). •• Explaining the tumor-associated factors, exosomes, contributing to NK cells-based immunotherapy subversion.
    • 88. Klingemann H-G, Wong E, Maki G. A cytotoxic NK-cell line (NK-92) for ex vivo purging of leukemia from blood. Bio. Blood Marrow Transplant. 2(2), 68–75 (1996).
    • 89. Klingemann H, Boissel L, Toneguzzo F. Natural killer cells for immunotherapy – advantages of the NK-92 cell line over blood NK cells. Front. Immunol. 7, 91 (2016). •• Potential utility of NK-92 cells for cancer immunotherapy.
    • 90. Lapteva N, Parihar R, Rollins LA, Gee AP, Rooney CM. Large-scale culture and genetic modification of human natural killer cells for cellular therapy. In: Natural Killer Cells. Springer, Clifton, NJ, USA, 195–202 (2016).
    • 91. Ruella M, Xu J, Barrett DM et al. Induction of resistance to chimeric antigen receptor T cell therapy by transduction of a single leukemic B cell. Nat. Med. 24(10), 1499–1503 (2018).
    • 92. Zhang C, Oberoi P, Oelsner S et al. Chimeric antigen receptor-engineered NK-92 cells: an off-the-shelf cellular therapeutic for targeted elimination of cancer cells and induction of protective antitumor immunity. Front. Immunol. 8, 533 (2017).
    • 93. Clémenceau B, Vivien R, Pellat C, Foss M, Thibault G, Vié H. The human natural killer cytotoxic cell line NK-92, once armed with a murine CD16 receptor, represents a convenient cellular tool for the screening of mouse mAbs according to their ADCC potential. In: MAbs. Taylor andFrancis, NY, USA, 587–594 (2013)
    • 94. Li F, Liu S. Focusing on NK cells and ADCC: a promising immunotherapy approach in targeted therapy for HER2-positive breast cancer. Front. Immunol. 13, doi: 10.3389/fimmu.2022.1083462 (2022) (Online).
    • 95. Zhang J, Zheng H, Diao Y. Natural killer cells and current applications of chimeric antigen receptor-modified NK-92 cells in tumor immunotherapy. Int. J. Mol. Sci. 20(2), 317 (2019).
    • 96. Sivori S, Meazza R, Quintarelli C et al. NK cell-based immunotherapy for hematological malignancies. J. Clin. Med. 8(10), 1702 (2019).
    • 97. Zhou Y, Li M, Zhou K et al. Engineering induced pluripotent stem cells for cancer immunotherapy. Cancers (Basel) 14(9), 2266 (2022).
    • 98. Crow D. Could iPSCs enable ‘off-the-shelf’ cell therapy? Cell 177(7), 1667–1669 (2019).
    • 99. Jung IH, Kim DH, Yoo DK et al. In vivo study of natural killer (NK) cell cytotoxicity against cholangiocarcinoma in a nude mouse model. In Vivo 32(4), 771 (2018).
    • 100. Dahlberg CI, Sarhan D, Chrobok M, Duru AD, Alici E. Natural killer cell-based therapies targeting cancer: possible strategies to gain and sustain anti-tumor activity. Front. Immunol. 6, 605 (2015).
    • 101. Burga RA, Nguyen T, Zulovich J et al. Improving efficacy of cancer immunotherapy by genetic modification of natural killer cells. Cytotherapy 18(11), 1410–1421 (2016).
    • 102. Murray S, Lundqvist A. Targeting the tumor microenvironment to improve natural killer cell-based immunotherapies: on being in the right place at the right time, with resilience. Hum. Vaccin. Immunother. 12(3), 607–611 (2016).
    • 103. Vitale M, Cantoni C, Pietra G, Mingari MC, Moretta L. Effect of tumor cells and tumor microenvironment on NK-cell function. Eur. J. Immunol. 44(6), 1582–1592 (2014).
    • 104. Ghiringhelli F, Ménard C, Terme M et al. CD4+ CD25+ regulatory T cells inhibit natural killer cell functions in a transforming growth factor–β–dependent manner. J. Exp. Med. 202(8), 1075–1085 (2005).
    • 105. Beavis PA, Divisekera U, Paget C et al. Blockade of A2A receptors potently suppresses the metastasis of CD73+ tumors. Proc. Natl Acad. Sci. USA 110(36), 14711–14716 (2013).
    • 106. Kai S, Goto S, Tahara K, Sasaki A, Kawano K, Kitano S. Inhibition of indoleamine 2, 3-dioxygenase suppresses NK cell activity and accelerates tumor growth. J. Exp. Ther. Oncol. 3(6), 336–345 (2003).
    • 107. Fauriat C, Just-Landi S, Mallet F et al. Deficient expression of NCR in NK cells from acute myeloid leukemia: evolution during leukemia treatment and impact of leukemia cells in NCRdull phenotype induction. Blood 109(1), 323–330 (2007).
    • 108. Sanchez-Correa B, Gayoso I, Bergua JM et al. Decreased expression of DNAM-1 on NK cells from acute myeloid leukemia patients. Immunol. Cell Biol. 90(1), 109–115 (2012).