Skip to main content

Thank you for visiting nature.com. You are using a browser version with limited support for CSS. To obtain the best experience, we recommend you use a more up to date browser (or turn off compatibility mode in Internet Explorer). In the meantime, to ensure continued support, we are displaying the site without styles and JavaScript.

  • Review Article
  • Published:

NK cells and cancer: you can teach innate cells new tricks

Key Points

  • Natural killer (NK) cells have a primordial role in tumour immunosurveillance. Given their potent antitumour activity, therapeutic manipulation of NK cells provides an attractive strategy for cancer treatment.

  • A balance of inhibitory and stimulatory signals delivered by numerous receptors regulates NK cell functions (cytotoxicity, cytokine production and proliferation). NKG2D is the best characterized of these activating receptors and is directly involved in the recognition of tumours that express NKG2D ligands.

  • NK cells detect and eliminate tumour cells that are deficient in major histocompatibility complex (MHC) class I molecules (missing self), have upregulated stress ligands that are recognized by activating NK cell receptors (induced self), or are coated with antibodies specific for tumour antigens, enabling recognition by NK cells expressing the CD16 Fc receptor, which triggers antibody-dependent cell-mediated cytotoxicity.

  • Immunoediting of tumour cells by NK cells leads to tumour variants that can evade NK cell-mediated responses. Direct and indirect tumour escape mechanisms include secretion of immunosuppressive factors, shedding of ligands for the activating NK cell receptors and induction of ligands for activating NK cell receptors on healthy cells that can serve as decoys.

  • Current checkpoint blockade agents used to activate T cells in cancer patients, such as antibodies against PD1, may also target NK cells. Checkpoint blockade targeting inhibitory receptors shared by NK cells and T cells are currently under clinical evaluation.

  • In addition to engineered T cells, NK cells and their receptors represent an attractive alternative for the use of chimeric antigen receptors.

  • Immunotherapeutic approaches such as the generation of bispecific linkers that engage activating NK cell receptors and tumour-specific antigens are also being developed to unleash the antitumour potential of NK cells.

Abstract

Natural killer (NK) cells are the prototype innate lymphoid cells endowed with potent cytolytic function that provide host defence against microbial infection and tumours. Here, we review evidence for the role of NK cells in immune surveillance against cancer and highlight new therapeutic approaches for targeting NK cells in the treatment of cancer.

This is a preview of subscription content, access via your institution

Access options

Rent or buy this article

Prices vary by article type

from$1.95

to$39.95

Prices may be subject to local taxes which are calculated during checkout

Figure 1: Schematic representation of physiological NK cell functions.
Figure 2: Schematic representations of immune escape mechanisms used by tumour cells.
Figure 3: Schematic representation of therapeutic approaches that take advantage of NK cell potential.
Figure 4: Schematic representation summarizing the different NK cell-based therapeutic approaches.

Similar content being viewed by others

References

  1. Artis, D. & Spits, H. The biology of innate lymphoid cells. Nature 517, 293–301 (2015).

    Article  CAS  PubMed  Google Scholar 

  2. Orange, J. S. Natural killer cell deficiency. J. Allergy Clin. Immunol. 132, 515–525 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  3. Lanier, L. L. Up on the tightrope: natural killer cell activation and inhibition. Nat. Immunol. 9, 495–502 (2008).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  4. Campbell, K. S. & Purdy, A. K. Structure/function of human killer cell immunoglobulin-like receptors: lessons from polymorphisms, evolution, crystal structures and mutations. Immunology 132, 315–325 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  5. Schenkel, A. R., Kingry, L. C. & Slayden, R. A. The Ly49 gene family. A brief guide to the nomenclature, genetics, and role in intracellular infection. Front. Immunol. 4, 90 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  6. Lazetic, S., Chang, C., Houchins, J. P., Lanier, L. L. & Phillips, J. H. Human natural killer cell receptors involved in MHC class I recognition are disulfide-linked heterodimers of CD94 and NKG2 subunits. J. Immunol. 157, 4741–4745 (1996).

    CAS  PubMed  Google Scholar 

  7. Wada, H., Matsumoto, N., Maenaka, K., Suzuki, K. & Yamamoto, K. The inhibitory NK cell receptor CD94/NKG2A and the activating receptor CD94/NKG2C bind the top of HLA-E through mostly shared but partly distinct sets of HLA-E residues. Eur. J. Immunol. 34, 81–90 (2004).

    Article  CAS  PubMed  Google Scholar 

  8. Braud, V. M. et al. HLA-E binds to natural killer cell receptors CD94/NKG2A, B and C. Nature 391, 795–799 (1998).

    Article  CAS  PubMed  Google Scholar 

  9. Lee, N. et al. HLA-E is a major ligand for the natural killer inhibitory receptor CD94/NKG2A. Proc. Natl Acad. Sci. USA 95, 5199–5204 (1998).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  10. Vance, R. E., Kraft, J. R., Altman, J. D., Jensen, P. E. & Raulet, D. H. Mouse CD94/NKG2A Is a natural killer cell receptor for the nonclassical major histocompatibility complex (MHC) Class I molecule Qa-1b. J. Exp. Med. 188, 1841–1848 (1998).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  11. Lopez-Vergès, S. et al. Expansion of a unique CD57+NKG2Chi natural killer cell subset during acute human cytomegalovirus infection. Proc. Natl Acad. Sci. USA 108, 14725–14732 (2011).

    Article  PubMed  PubMed Central  Google Scholar 

  12. Cassidy, S. A., Cheent, K. S. & Khakoo, S. I. Effects of peptide on NK cell-mediated MHC I recognition. Front. Immunol. 5, 133 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  13. Tripathy, S. K., Smith, H. R. C., Holroyd, E. A., Pingel, J. T. & Yokoyama, W. M. Expression of m157, a murine cytomegalovirus-encoded putative major histocompatibility class I (MHC-I)-like protein, is independent of viral regulation of host MHC-I. J. Virol. 80, 545–550 (2006).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  14. Vivier, E. et al. Innate or adaptive immunity? The example of natural killer cells. Science 331, 44–49 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  15. Bauer, S. et al. Activation of NK cells and T cells by NKG2D, a receptor for stress-inducible MICA. Science 285, 727–729 (1999).

    Article  CAS  PubMed  Google Scholar 

  16. Lanier, L. L. NKG2D receptor and its ligands in host defense. Cancer Immunol. Res. 3, 575–582 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  17. Raulet, D. H., Gasser, S., Gowen, B. G., Deng, W. & Jung, H. Regulation of ligands for the NKG2D activating receptor. Annu. Rev. Immunol. 31, 413–441 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  18. Kruse, P. H., Matta, J., Ugolini, S. & Vivier, E. Natural cytotoxicity receptors and their ligands. Immunol. Cell Biol. 92, 221–229 (2014).

    Article  CAS  PubMed  Google Scholar 

  19. Pende, D. et al. Identification and molecular characterization of Nkp30, a novel triggering receptor involved in natural cytotoxicity mediated by human natural killer cells. J. Exp. Med. 190, 1505–1516 (1999).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  20. Pessino, A. et al. Molecular cloning of NKp46: a novel member of the immunoglobulin superfamily involved in triggering of natural cytotoxicity. J. Exp. Med. 188, 953–960 (1998).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  21. Cantoni, C. et al. NKp44, a triggering receptor involved in tumor cell lysis by activated human natural killer cells, is a novel member of the immunoglobulin superfamily. J. Exp. Med. 189, 787–796 (1999).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  22. Walzer, T. et al. Identification, activation, and selective in vivo ablation of mouse NK cells via NKp46. Proc. Natl Acad. Sci. USA 104, 3384–3389 (2007).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  23. Hollyoake, M., Campbell, R. D. & Aguado, B. NKp30 (NCR3) is a pseudogene in 12 inbred and wild mouse strains, but an expressed gene in Mus caroli. Mol. Biol. Evol. 22, 1661–1672 (2005).

    Article  CAS  PubMed  Google Scholar 

  24. Martinet, L. & Smyth, M. J. Balancing natural killer cell activation through paired receptors. Nat. Rev. Immunol. 15, 243–254 (2015).

    Article  CAS  PubMed  Google Scholar 

  25. Lakshmikanth, T. et al. NCRs and DNAM-1 mediate NK cell recognition and lysis of human and mouse melanoma cell lines in vitro and in vivo. J. Clin. Invest. 119, 1251–1263 (2009).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  26. Iguchi-Manaka, A. et al. Accelerated tumor growth in mice deficient in DNAM-1 receptor. J. Exp. Med. 205, 2959–2964 (2008). This article provides evidence that the DNAM1 receptor has an important role in immune surveillance of tumour development.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  27. Gilfillan, S. et al. DNAM-1 promotes activation of cytotoxic lymphocytes by nonprofessional antigen-presenting cells and tumors. J. Exp. Med. 205, 2965–2973 (2008).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  28. Chan, C. J. et al. The receptors CD96 and CD226 oppose each other in the regulation of natural killer cell functions. Nat. Immunol. 15, 431–438 (2014).

    Article  CAS  PubMed  Google Scholar 

  29. Johnston, R. J. et al. The immunoreceptor TIGIT regulates antitumor and antiviral CD8+ T cell effector function. Cancer Cell 26, 923–937 (2014). References 28 and 29 demonstrate the role of DNAM1, CD96 and TIGIT in the regulation of NK cell and T cell responses against tumours.

    Article  CAS  PubMed  Google Scholar 

  30. Bruhns, P. Properties of mouse and human IgG receptors and their contribution to disease models. Blood 119, 5640–5649 (2012).

    Article  CAS  PubMed  Google Scholar 

  31. Battella, S., Cox, M. C., Santoni, A. & Palmieri, G. Natural killer (NK) cells and anti-tumor therapeutic mAb: unexplored interactions. J. Leukoc. Biol., http://dx.doi.org/10.1189/jlb.5VMR0415-141R (2015).

  32. Voskoboinik, I., Whisstock, J. C. & Trapani, J. A. Perforin and granzymes: function, dysfunction and human pathology. Nat. Rev. Immunol. 15, 388–400 (2015).

    Article  CAS  PubMed  Google Scholar 

  33. Krzewski, K. & Coligan, J. E. Human NK cell lytic granules and regulation of their exocytosis. NK Cell Biol. 3, 335 (2012).

    Google Scholar 

  34. Wallin, R. P. A., Screpanti, V., Michaëlsson, J., Grandien, A. & Ljunggren, H.-G. Regulation of perforin-independent NK cell-mediated cytotoxicity. Eur. J. Immunol. 33, 2727–2735 (2003).

    Article  CAS  PubMed  Google Scholar 

  35. Smyth, M. J. et al. Activation of NK cell cytotoxicity. Mol. Immunol. 42, 501–510 (2005).

    Article  CAS  PubMed  Google Scholar 

  36. Cretney, E. et al. Increased susceptibility to tumor initiation and metastasis in TNF-related apoptosis-inducing ligand-deficient mice. J. Immunol. 168, 1356–1361 (2002).

    Article  CAS  PubMed  Google Scholar 

  37. Cooper, M. A. et al. Human natural killer cells: a unique innate immunoregulatory role for the CD56bright subset. Blood 97, 3146–3151 (2001).

    Article  CAS  PubMed  Google Scholar 

  38. Maghazachi, A. A. in The Chemokine System in Experimental and Clinical Hematology (ed. Bruserud, O.) 37–58 (Springer, 2010).

    Book  Google Scholar 

  39. O'Sullivan, T. et al. Cancer immunoediting by the innate immune system in the absence of adaptive immunity. J. Exp. Med. 209, 1869–1882 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  40. Roder, J. C. et al. A new immunodeficiency disorder in humans involving NK cells. Nature 284, 553–555 (1980).

    Article  CAS  PubMed  Google Scholar 

  41. Sullivan, J. L., Byron, K. S., Brewster, F. E. & Purtilo, D. T. Deficient natural killer cell activity in x-linked lymphoproliferative syndrome. Science 210, 543–545 (1980).

    Article  CAS  PubMed  Google Scholar 

  42. Talmadge, J. E., Meyers, K. M., Prieur, D. J. & Starkey, J. R. Role of natural killer cells in tumor growth and metastasis: C57BL/6 normal and beige mice. J. Natl Cancer Inst. 65, 929–935 (1980).

    CAS  PubMed  Google Scholar 

  43. Gorelik, E., Wiltrout, R. H., Okumura, K., Habu, S. & Herberman, R. B. Role of NK cells in the control of metastatic spread and growth of tumor cells in mice. Int. J. Cancer 30, 107–112 (1982).

    Article  CAS  PubMed  Google Scholar 

  44. Nakajima, T., Mizushima, N., Nakamura, J. & Kanai, K. Surface markers of NK cells in peripheral blood of patients with cirrhosis and hepatocellular carcinoma. Immunol. Lett. 13, 7–10 (1986).

    Article  CAS  PubMed  Google Scholar 

  45. Pross, H. F. & Lotzová, E. Role of natural killer cells in cancer. Nat. Immunol. 12, 279–292 (1993).

    CAS  Google Scholar 

  46. Schantz, S. P., Shillitoe, E. J., Brown, B. & Campbell, B. Natural killer cell activity and head and neck cancer: a clinical assessment. J. Natl Cancer Inst. 77, 869–875 (1986).

    CAS  PubMed  Google Scholar 

  47. Strayer, D. R., Carter, W. A. & Brodsky, I. Familial occurrence of breast cancer is associated with reduced natural killer cytotoxicity. Breast Cancer Res. Treat. 7, 187–192 (1986).

    Article  CAS  PubMed  Google Scholar 

  48. Hersey, P., Edwards, A., Honeyman, M. & McCarthy, W. H. Low natural-killer-cell activity in familial melanoma patients and their relatives. Br. J. Cancer 40, 113–122 (1979).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  49. Imai, K., Matsuyama, S., Miyake, S., Suga, K. & Nakachi, K. Natural cytotoxic activity of peripheral-blood lymphocytes and cancer incidence: an 11-year follow-up study of a general population. Lancet 356, 1795–1799 (2000). This is a clinical study correlating susceptibility to cancer and NK cell function.

    Article  CAS  PubMed  Google Scholar 

  50. Spinner, M. A. et al. GATA2 deficiency: a protean disorder of hematopoiesis, lymphatics and immunity. Blood 123, 809–821 (2013).

    Article  CAS  PubMed  Google Scholar 

  51. Gineau, L. et al. Partial MCM4 deficiency in patients with growth retardation, adrenal insufficiency, and natural killer cell deficiency. J. Clin. Invest. 122, 821–832 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  52. Smyth, M. J., Hayakawa, Y., Takeda, K. & Yagita, H. New aspects of natural-killer-cell surveillance and therapy of cancer. Nat. Rev. Cancer 2, 850–861 (2002).

    Article  CAS  PubMed  Google Scholar 

  53. Bezman, N. A. et al. Molecular definition of the identity and activation of natural killer cells. Nat. Immunol. 13, 1000–1009 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  54. Hudspeth, K., Silva-Santos, B. & Mavilio, D. Natural cytotoxicity receptors: broader expression patterns and functions in innate and adaptive immune cells. NK Cell Biol. 4, 69 (2013).

    Google Scholar 

  55. Robinette, M. L. et al. Transcriptional programs define molecular characteristics of innate lymphoid cell classes and subsets. Nat. Immunol. 16, 306–317 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  56. Albertsson, P. A. et al. NK cells and the tumour microenvironment: implications for NK-cell function and anti-tumour activity. Trends Immunol. 24, 603–609 (2003).

    Article  CAS  PubMed  Google Scholar 

  57. Bix, M. et al. Rejection of class I MHC-deficient haemopoietic cells by irradiated MHC-matched mice. Nature 349, 329–331 (1991).

    Article  CAS  PubMed  Google Scholar 

  58. Nausch, N. & Cerwenka, A. NKG2D ligands in tumor immunity. Oncogene 27, 5944–5958 (2008).

    Article  CAS  PubMed  Google Scholar 

  59. Cerwenka, A., Baron, J. L. & Lanier, L. L. Ectopic expression of retinoic acid early inducible-1 gene (RAE-1) permits natural killer cell-mediated rejection of a MHC class I-bearing tumor in vivo. Proc. Natl Acad. Sci. USA 98, 11521–11526 (2001).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  60. Diefenbach, A., Jensen, E. R., Jamieson, A. M. & Raulet, D. H. Rae1 and H60 ligands of the NKG2D receptor stimulate tumour immunity. Nature 413, 165–171 (2001).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  61. Guerra, N. et al. NKG2D-deficient mice are defective in tumor surveillance in models of spontaneous malignancy. Immunity 28, 571–580 (2008). This article provides evidence for the role of NKG2D in tumour immunosurveillance in mouse tumour models.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  62. Hayakawa, Y. et al. Cutting edge: tumor rejection mediated by NKG2D receptor–ligand interaction is dependent upon perforin. J. Immunol. 169, 5377–5381 (2002).

    Article  CAS  PubMed  Google Scholar 

  63. Groh, V., Wu, J., Yee, C. & Spies, T. Tumour-derived soluble MIC ligands impair expression of NKG2D and T-cell activation. Nature 419, 734–738 (2002).

    Article  CAS  PubMed  Google Scholar 

  64. Kaiser, B. K. et al. Disulphide-isomerase-enabled shedding of tumour-associated NKG2D ligands. Nature 447, 482–486 (2007).

    Article  CAS  PubMed  Google Scholar 

  65. Salih, H. R., Rammensee, H.-G. & Steinle, A. Cutting edge: down-regulation of MICA on human tumors by proteolytic shedding. J. Immunol. 169, 4098–4102 (2002).

    Article  CAS  PubMed  Google Scholar 

  66. Wu, J. D. et al. Prevalent expression of the immunostimulatory MHC class I chain–related molecule is counteracted by shedding in prostate cancer. J. Clin. Invest. 114, 560–568 (2004). References 63–66 describe mechanisms by which tumours shed NKG2DLs to avoid NK cell and T cell recognition.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  67. Fernández-Messina, L. et al. Differential mechanisms of shedding of the glycosylphosphatidylinositol (GPI)-anchored NKG2D ligands. J. Biol. Chem. 285, 8543–8551 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  68. Salih, H. R., Goehlsdorf, D. & Steinle, A. Release of MICB molecules by tumor cells: mechanism and soluble MICB in sera of cancer patients. Hum. Immunol. 67, 188–195 (2006).

    Article  CAS  PubMed  Google Scholar 

  69. Salih, H. R., Holdenrieder, S. & Steinle, A. Soluble NKG2D ligands: prevalence, release, and functional impact. Front. Biosci. 13, 3448–3456 (2008).

    Article  CAS  PubMed  Google Scholar 

  70. Holdenrieder, S. et al. Soluble MICA in malignant diseases. Int. J. Cancer 118, 684–687 (2006).

    Article  CAS  PubMed  Google Scholar 

  71. Crane, C. A. et al. Immune evasion mediated by tumor-derived lactate dehydrogenase induction of NKG2D ligands on myeloid cells in glioblastoma patients. Proc. Natl Acad. Sci. USA 111, 12823–12828 (2014). This article shows that the secretion of lactate dehydrogenase by human glioblastomas induces NKG2D ligands on monocytes, which may serve as decoys to evade antitumour immune responses.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  72. Champsaur, M. & Lanier, L. L. Effect of NKG2D ligand expression on host immune responses. Immunol. Rev. 235, 267–285 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  73. Oppenheim, D. E. et al. Sustained localized expression of ligand for the activating NKG2D receptor impairs natural cytotoxicity in vivo and reduces tumor immunosurveillance. Nat. Immunol. 6, 928–937 (2005).

    Article  CAS  PubMed  Google Scholar 

  74. Deng, W. et al. A shed NKG2D ligand that promotes natural killer cell activation and tumor rejection. Science 348, 136–139 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  75. Kopp, H.-G., Placke, T. & Salih, H. R. Platelet-derived transforming growth factor-β down-regulates NKG2D thereby inhibiting natural killer cell antitumor reactivity. Cancer Res. 69, 7775–7783 (2009).

    Article  CAS  PubMed  Google Scholar 

  76. Placke, T., Kopp, H.-G. & Salih, H. R. The wolf in sheep's clothing. Oncoimmunology 1, 557–559 (2012).

    Article  PubMed  PubMed Central  Google Scholar 

  77. Placke, T., Salih, H. R. & Kopp, H.-G. GITR ligand provided by thrombopoietic cells inhibits NK cell antitumor activity. J. Immunol. 189, 154–160 (2012).

    Article  CAS  PubMed  Google Scholar 

  78. Placke, T., Kopp, H.-G. & Salih, H. R. Modulation of natural killer cell anti-tumor reactivity by platelets. J. Innate Immun. 3, 374–382 (2011).

    Article  CAS  PubMed  Google Scholar 

  79. Wilson, E. B. et al. Human tumour immune evasion via TGF-β blocks NK cell activation but not survival allowing therapeutic restoration of anti-tumour activity. PLoS ONE 6, e22842 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  80. Holt, D., Ma, X., Kundu, N. & Fulton, A. Prostaglandin E2 (PGE2) suppresses natural killer cell function primarily through the PGE2 receptor EP4. Cancer Immunol. Immunother. 60, 1577–1586 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  81. Pietra, G. et al. Melanoma cells inhibit natural killer cell function by modulating the expression of activating receptors and cytolytic activity. Cancer Res. 72, 1407–1415 (2012).

    Article  CAS  PubMed  Google Scholar 

  82. Hoskin, D., Mader, J., Furlong, S., Conrad, D. & Blay, J. Inhibition of T cell and natural killer cell function by adenosine and its contribution to immune evasion by tumor cells (Review). Int. J. Oncol. 32, 527–535 (2008).

    CAS  PubMed  Google Scholar 

  83. Richards, J. O. et al. Tumor growth impedes natural-killer-cell maturation in the bone marrow. Blood 108, 246–252 (2006).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  84. Mocellin, S., Marincola, F. M. & Young, H. A. Interleukin-10 and the immune response against cancer: a counterpoint. J. Leukoc. Biol. 78, 1043–1051 (2005).

    Article  CAS  PubMed  Google Scholar 

  85. Park, J. Y. et al. IL-15-induced IL-10 increases the cytolytic activity of human natural killer cells. Mol. Cells 32, 265–272 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  86. Moretta, A. et al. Activating receptors and coreceptors involved in human natural killer cell-mediated cytolysis. Annu. Rev. Immunol. 19, 197–223 (2001).

    Article  CAS  PubMed  Google Scholar 

  87. Byrd, A., Hoffmann, S. C., Jarahian, M., Momburg, F. & Watzl, C. Expression analysis of the ligands for the natural killer cell receptors NKp30 and NKp44. PLoS ONE 2, e1339 (2007).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  88. Cagnano, E. et al. Expression of ligands to NKp46 in benign and malignant melanocytes. J. Invest. Dermatol. 128, 972–979 (2007).

    Article  CAS  PubMed  Google Scholar 

  89. Brandt, C. S. et al. The B7 family member B7-H6 is a tumor cell ligand for the activating natural killer cell receptor NKp30 in humans. J. Exp. Med. 206, 1495–1503 (2009).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  90. Li, Y., Wang, Q. & Mariuzza, R. A. Structure of the human activating natural cytotoxicity receptor NKp30 bound to its tumor cell ligand B7-H6. J. Exp. Med. 208, 703–714 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  91. Matta, J. et al. Induction of B7-H6, a ligand for the natural killer cell–activating receptor NKp30, in inflammatory conditions. Blood 122, 394–404 (2013).

    Article  CAS  PubMed  Google Scholar 

  92. Halfteck, G. G. et al. Enhanced in vivo growth of lymphoma tumors in the absence of the NK-activating receptor NKp46/NCR1. J. Immunol. 182, 2221–2230 (2009).

    Article  CAS  PubMed  Google Scholar 

  93. Glasner, A. et al. Recognition and prevention of tumor metastasis by the NK receptor NKp46/NCR1. J. Immunol. 188, 2509–2515 (2012).

    Article  CAS  PubMed  Google Scholar 

  94. Alvarez-Breckenridge, C. A. et al. NK cells impede glioblastoma virotherapy through NKp30 and NKp46 natural cytotoxicity receptors. Nat. Med. 18, 1827–1834 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  95. Schreiber, R. D., Old, L. J. & Smyth, M. J. Cancer immunoediting: integrating immunity's roles in cancer suppression and promotion. Science 331, 1565–1570 (2011).

    Article  CAS  PubMed  Google Scholar 

  96. Smyth, M. J. et al. NKG2D function protects the host from tumor initiation. J. Exp. Med. 202, 583–588 (2005).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  97. Elboim, M. et al. Tumor immunoediting by NKp46. J. Immunol. 184, 5637–5644 (2010).

    Article  CAS  PubMed  Google Scholar 

  98. Alvarez, I. B. et al. Role played by the programmed death-1-programmed death ligand pathway during innate immunity against Mycobacterium tuberculosis. J. Infect. Dis. 202, 524–532 (2010).

    Article  CAS  PubMed  Google Scholar 

  99. Norris, S. et al. PD-1 expression on natural killer cells and CD8+ T cells during chronic HIV-1 infection. Viral Immunol. 25, 329–332 (2012).

    Article  CAS  PubMed  Google Scholar 

  100. Stojanovic, A., Fiegler, N., Brunner-Weinzierl, M. & Cerwenka, A. CTLA-4 is expressed by activated mouse NK cells and inhibits NK cell IFN-γ production in response to mature dendritic cells. J. Immunol. 192, 4184–4191 (2014).

    Article  CAS  PubMed  Google Scholar 

  101. Benson, D. M. et al. The PD-1/PD-L1 axis modulates the natural killer cell versus multiple myeloma effect: a therapeutic target for CT-011, a novel monoclonal anti–PD-1 antibody. Blood 116, 2286–2294 (2010). This is a report of the expression of PD1 on NK cells and augmentation of NK cell activity against multiple myeloma by blockade of PD1.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  102. Boyerinas, B. et al. Antibody dependent cellular cytotoxicity activity of a novel anti-PD-L1 antibody, avelumab (MSB0010718C), on human tumor cells. Cancer Immunol. Res. 3, 1148–1157 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  103. Lang, S., Vujanovic, N. L., Wollenberg, B. & Whiteside, T. L. Absence of B7.1-CD28/CTLA-4-mediated co-stimulation in human NK cells. Eur. J. Immunol. 28, 780–786 (1998).

    Article  CAS  PubMed  Google Scholar 

  104. Chambers, B. J., Salcedo, M. & Ljunggren, H.-G. Triggering of natural killer cells by the costimulatory molecule CD80 (B7-1). Immunity 5, 311–317 (1996).

    Article  CAS  PubMed  Google Scholar 

  105. Contardi, E. et al. CTLA-4 is constitutively expressed on tumor cells and can trigger apoptosis upon ligand interaction. Int. J. Cancer 117, 538–550 (2005).

    Article  CAS  PubMed  Google Scholar 

  106. Laurent, S. et al. The engagement of CTLA-4 on primary melanoma cell lines induces antibody-dependent cellular cytotoxicity and TNF-α production. J. Transl Med. 11, 108 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  107. Jarahian, M. et al. Activation of natural killer cells by Newcastle disease virus hemagglutinin-neuraminidase. J. Virol. 83, 8108–8121 (2009).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  108. Bhat, R., Dempe, S., Dinsart, C. & Rommelaere, J. Enhancement of NK cell antitumor responses using an oncolytic parvovirus. Int. J. Cancer 128, 908–919 (2011).

    Article  CAS  PubMed  Google Scholar 

  109. Zamarin, D. et al. Localized oncolytic virotherapy overcomes systemic tumor resistance to immune checkpoint blockade immunotherapy. Sci. Transl Med. 6, 226ra32 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  110. Romagné, F. et al. Preclinical characterization of 1-7F9, a novel human anti–KIR receptor therapeutic antibody that augments natural killer–mediated killing of tumor cells. Blood 114, 2667–2677 (2009).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  111. Vahlne, G. et al. In vivo tumor cell rejection induced by NK cell inhibitory receptor blockade: maintained tolerance to normal cells even in the presence of IL-2. Eur. J. Immunol. 40, 813–823 (2010).

    Article  CAS  PubMed  Google Scholar 

  112. Kohrt, H. E. et al. Anti-KIR antibody enhancement of anti-lymphoma activity of natural killer cells as monotherapy and in combination with anti-CD20 antibodies. Blood 123, 678–686 (2014). This article reports that KIR blockade enhances the ADCC activity of NK cells in preclinical models.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  113. Vey, N. et al. A phase 1 trial of the anti-inhibitory KIR mAb IPH2101 for AML in complete remission. Blood 120, 4317–4323 (2012).

    Article  CAS  PubMed  Google Scholar 

  114. Benson, D. M. et al. A Phase I trial of the anti-KIR antibody IPH2101 and lenalidomide in patients with relapsed/refractory multiple myeloma. Clin. Cancer Res. 21, 4055–4061 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  115. Benson, D. M. et al. A phase 1 trial of the anti-KIR antibody IPH2101 in patients with relapsed/refractory multiple myeloma. Blood 120, 4324–4333 (2012). References 113–115 report Phase I and II trials with KIR blockade in cancer patients.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  116. Seymour, L., Tinker, A., Hirte, H., Wagtmann, N. & Dodion, P. Abstract O3.2. Phase I and dose ranging, phase II studies with IPH2201, a humanized monoclonal antibody targeting HLA-E receptor CD94/NKG2A. Ann. Oncol. 26(Suppl 2), ii3–ii3 (2015).

    Article  Google Scholar 

  117. Ndhlovu, L. C. et al. Tim-3 marks human natural killer cell maturation and suppresses cell-mediated cytotoxicity. Blood 119, 3734–3743 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  118. Sun, H., Sun, C. & Xiao, W. Expression regulation of co-inhibitory molecules on human natural killer cells in response to cytokine stimulations. Cytokine 65, 33–41 (2014).

    Article  CAS  PubMed  Google Scholar 

  119. Jing, W. et al. Combined immune checkpoint protein blockade and low dose whole body irradiation as immunotherapy for myeloma. J. Immunother. Cancer 3, 2 (2015).

    Article  PubMed  PubMed Central  Google Scholar 

  120. Ngiow, S. F., Teng, M. W. L. & Smyth, M. J. Prospects for TIM3-targeted antitumor immunotherapy. Cancer Res. 71, 6567–6571 (2011).

    Article  CAS  PubMed  Google Scholar 

  121. Woo, S.-R. et al. Immune inhibitory molecules LAG-3 and PD-1 synergistically regulate T-cell function to promote tumoral immune escape. Cancer Res. 72, 917–927 (2012).

    Article  CAS  PubMed  Google Scholar 

  122. Bartkowiak, T. & Curran, M. A. 4-1BB agonists: multi-potent potentiators of tumor immunity. Front. Oncol. 5, 117 (2015).

    Article  PubMed  PubMed Central  Google Scholar 

  123. Baessler, T. et al. CD137 ligand mediates opposite effects in human and mouse NK cells and impairs NK-cell reactivity against human acute myeloid leukemia cells. Blood 115, 3058–3069 (2010).

    Article  CAS  PubMed  Google Scholar 

  124. Kohrt, H. E. et al. Stimulation of natural killer cells with a CD137-specific antibody enhances trastuzumab efficacy in xenotransplant models of breast cancer. J. Clin. Invest. 122, 1066–1075 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  125. Kohrt, H. E. et al. CD137 stimulation enhances the antilymphoma activity of anti-CD20 antibodies. Blood 117, 2423–2432 (2011). References 124 and 125 demonstrate that agonist CD137 antibodies enhance NK cell-mediated ADCC in preclinical models.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  126. Ascierto, P. A., Simeone, E., Sznol, M., Fu, Y.-X. & Melero, I. Clinical experiences with anti-CD137 and anti-PD1 therapeutic antibodies. Semin. Oncol. 37, 508–516 (2010).

    Article  CAS  PubMed  Google Scholar 

  127. Melero, I., Hirschhorn-Cymerman, D., Morales-Kastresana, A., Sanmamed, M. F. & Wolchok, J. D. Agonist antibodies to TNFR molecules that costimulate T and NK cells. Clin. Cancer Res. 19, 1044–1053 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  128. Rosenberg, S. A. et al. Observations on the systemic administration of autologous lymphokine-activated killer cells and recombinant interleukin-2 to patients with metastatic cancer. N. Engl. J. Med. 313, 1485–1492 (1985).

    Article  CAS  PubMed  Google Scholar 

  129. Ruggeri, L. et al. Effectiveness of donor natural killer cell alloreactivity in mismatched hematopoietic transplants. Science 295, 2097–2100 (2002). This article supplies the first evidence that KIR-mismatched NK cells decrease leukaemia relapse in human patients receiving haematopoietic stem cell transplants.

    Article  CAS  PubMed  Google Scholar 

  130. Miller, J. S. et al. Successful adoptive transfer and in vivo expansion of human haploidentical NK cells in patients with cancer. Blood 105, 3051–3057 (2005). This article reports on the adoptive transfer of allogeneic NK cells in patients with leukaemia.

    Article  CAS  PubMed  Google Scholar 

  131. Ishikawa, E. et al. Autologous natural killer cell therapy for human recurrent malignant glioma. Anticancer Res. 24, 1861–1871 (2004).

    PubMed  Google Scholar 

  132. Shah, N. N. et al. Acute GVHD in patients receiving IL-15/4-1BBL activated NK cells following T-cell–depleted stem cell transplantation. Blood 125, 784–792 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  133. Cooper, M. A. et al. Cytokine-induced memory-like natural killer cells. Proc. Natl Acad. Sci. USA 106, 1915–1919 (2009).

    Article  PubMed  PubMed Central  Google Scholar 

  134. Ni, J., Miller, M., Stojanovic, A. & Cerwenka, A. Toward the next generation of NK cell-based adoptive cancer immunotherapy. Oncoimmunology 2 (2013).

  135. Ni, J., Miller, M., Stojanovic, A., Garbi, N. & Cerwenka, A. Sustained effector function of IL-12/15/18–preactivated NK cells against established tumors. J. Exp. Med. 209, 2351–2365 (2012). This article reports that adoptive transfer of cytokine-activated mouse NK cells shows efficacy against tumours in vivo.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  136. Ardolino, M., Hsu, J. & Raulet, D. H. Cytokine treatment in cancer immunotherapy. Oncotarget 6, 19346–19347 (2015).

    Article  PubMed  PubMed Central  Google Scholar 

  137. Ardolino, M. et al. Cytokine therapy reverses NK cell anergy in MHC-deficient tumors. J. Clin. Invest. 124, 4781–4794 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  138. Floros, T. & Tarhini, A. A. Anticancer cytokines: biology and clinical effects of interferon-α2, interleukin (IL)-2, IL-15, IL-21, and IL-12. Semin. Oncol. 42, 539–548 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  139. Guo, Y. et al. IL-15 superagonist–mediated immunotoxicity: role of NK cells and IFN-γ. J. Immunol. 195, 2353–2364 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  140. Glienke, W. et al. Advantages and applications of CAR-expressing natural killer cells. Exp. Pharmacol. Drug Discov. 6, 21 (2015).

    Google Scholar 

  141. Hermanson, D. L. & Kaufman, D. S. Utilizing chimeric antigen receptors to direct natural killer cell activity. Front. Immunol. 6, 195 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  142. Boissel, L., Betancur, M., Wels, W. S., Tuncer, H. & Klingemann, H. Transfection with mRNA for CD19 specific chimeric antigen receptor restores NK cell mediated killing of CLL cells. Leuk. Res. 33, 1255–1259 (2009).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  143. Müller, T. et al. Expression of a CD20-specific chimeric antigen receptor enhances cytotoxic activity of NK cells and overcomes NK-resistance of lymphoma and leukemia cells. Cancer Immunol. Immunother. 57, 411–423 (2007).

    Article  CAS  PubMed  Google Scholar 

  144. Esser, R. et al. NK cells engineered to express a GD2-specific antigen receptor display built-in ADCC-like activity against tumour cells of neuroectodermal origin. J. Cell. Mol. Med. 16, 569–581 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  145. Ahmed, M. & Cheung, N.-K. V. Engineering anti-GD2 monoclonal antibodies for cancer immunotherapy. FEBS Lett. 588, 288–297 (2014).

    Article  CAS  PubMed  Google Scholar 

  146. Uherek, C. et al. Retargeting of natural killer-cell cytolytic activity to ErbB2-expressing cancer cells results in efficient and selective tumor cell destruction. Blood 100, 1265–1273 (2002).

    CAS  PubMed  Google Scholar 

  147. Liu, H. et al. Specific growth inhibition of ErbB2-expressing human breast cancer cells by genetically modified NK92 cells. Oncol. Rep. 33, 95–102 (2015).

    CAS  PubMed  Google Scholar 

  148. Schönfeld, K. et al. Selective inhibition of tumor growth by clonal NK cells expressing an ErbB2/HER2-specific chimeric antigen receptor. Mol. Ther. 23, 330–338 (2015).

    Article  CAS  PubMed  Google Scholar 

  149. Sahm, C., Schönfeld, K. & Wels, W. S. Expression of IL-15 in NK cells results in rapid enrichment and selective cytotoxicity of gene-modified effectors that carry a tumor-specific antigen receptor. Cancer Immunol. Immunother. 61, 1451–1461 (2012).

    Article  CAS  PubMed  Google Scholar 

  150. Zhang, G. et al. Retargeting NK-92 for anti-melanoma activity by a TCR-like single-domain antibody. Immunol. Cell Biol. 91, 615–624 (2013).

    Article  CAS  PubMed  Google Scholar 

  151. Töpfer, K. et al. DAP12-based activating chimeric antigen receptor for NK cell tumor immunotherapy. J. Immunol. 194, 3201–3212 (2015).

    Article  CAS  PubMed  Google Scholar 

  152. Jiang, H. et al. Transfection of chimeric anti-CD138 gene enhances natural killer cell activation and killing of multiple myeloma cells. Mol. Oncol. 8, 297–310 (2014).

    Article  CAS  PubMed  Google Scholar 

  153. Knorr, D. A. & Kaufman, D. S. Pluripotent stem cell-derived natural killer cells for cancer therapy. Transl. Res. J. Lab. Clin. Med. 156, 147–154 (2010).

    Article  CAS  Google Scholar 

  154. Eguizabal, C. et al. Natural killer cells for cancer immunotherapy: pluripotent stem cells-derived NK cells as an immunotherapeutic perspective. NK Cell Biol. 5, 439 (2014).

    Google Scholar 

  155. Chu, J. et al. CS1-specific chimeric antigen receptor (CAR)-engineered natural killer cells enhance in vitro and in vivo antitumor activity against human multiple myeloma. Leukemia 28, 917–927 (2014).

    Article  CAS  PubMed  Google Scholar 

  156. Ni, Z. et al. Functional chimeric antigen receptor-expressing natural killer cells derived from human pluripotent stem cells. Blood 122, 896–896 (2013).

    Google Scholar 

  157. 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, 376–383 (2005).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  158. Li, L. et al. Expression of chimeric antigen receptors in natural killer cells with a regulatory-compliant non-viral method. Cancer Gene Ther. 17, 147–154 (2009).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  159. Shimasaki, N. et al. A clinically adaptable method to enhance the cytotoxicity of natural killer cells against B-cell malignancies. Cytotherapy 14, 830–840 (2012).

    Article  CAS  PubMed  Google Scholar 

  160. Alsamah, W. & Romia, Y. Modification of natural killer cells to target tumors. Int. J. Pharm. Clin. Res. 6, 97–100 (2014).

    Google Scholar 

  161. Altvater, B. et al. 2B4 (CD244) signaling by recombinant antigen-specific chimeric receptors costimulates natural killer cell activation to leukemia and neuroblastoma cells. Clin. Cancer Res. 15, 4857–4866 (2009).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  162. Müller, N. et al. Engineering NK cells modified with an EGFRvIII-specific chimeric antigen receptor to overexpress CXCR4 improves immunotherapy of CXCL12/SDF-1α-secreting glioblastoma. J. Immunother. 38, 197–210 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  163. Han, J. 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, 11483 (2015).

    Article  PubMed  PubMed Central  Google Scholar 

  164. Boissel, L. et al. Retargeting NK-92 cells by means of CD19- and CD20-specific chimeric antigen receptors compares favorably with antibody-dependent cellular cytotoxicity. Oncoimmunology 2 (2013).

  165. Klingemann, H. Are natural killer cells superior CAR drivers? OncoImmunology 3, e28147 (2014).

    Article  PubMed  PubMed Central  Google Scholar 

  166. Sentman, C. L. & Meehan, K. R. NKG2D CARs as cell therapy for cancer. Cancer J. 20, 156–159 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  167. Barber, A., Rynda, A. & Sentman, C. L. Chimeric NKG2D expressing T cells eliminate immunosuppression and activate immunity within the ovarian tumor microenvironment. J. Immunol. 183, 6939–6947 (2009).

    Article  CAS  PubMed  Google Scholar 

  168. Roy, S. et al. NK cells lyse T regulatory cells that expand in response to an intracellular pathogen. J. Immunol. 180, 1729–1736 (2008).

    Article  CAS  PubMed  Google Scholar 

  169. Zhang, T. & Sentman, C. L. Cancer immunotherapy using a bispecific NK receptor fusion protein that engages both T cells and tumor cells. Cancer Res. 71, 2066–2076 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  170. Lehner, M. et al. Redirecting T cells to Ewing's sarcoma family of tumors by a chimeric NKG2D receptor expressed by lentiviral transduction or mRNA transfection. PLoS ONE 7, e31210 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  171. VanSeggelen, H. et al. T cells engineered with chimeric antigen receptors targeting NKG2D ligands display lethal toxicity in mice. Mol. Ther. http://dx.doi.org/10.1038/mt.2015.119 (2015).

  172. Kudo, K. et al. T lymphocytes expressing a CD16 signaling receptor exert antibody-dependent cancer cell killing. Cancer Res. 74, 93–103 (2014).

    Article  CAS  PubMed  Google Scholar 

  173. Tal, Y. et al. An NCR1-based chimeric receptor endows T-cells with multiple anti-tumor specificities. Oncotarget 5, 10949–10958 (2014).

    Article  PubMed  PubMed Central  Google Scholar 

  174. Zhang, T., Wu, M.-R. & Sentman, C. L. An NKp30-based chimeric antigen receptor promotes T cell effector functions and antitumor efficacy in vivo. J. Immunol. 189, 2290–2299 (2012).

    Article  CAS  PubMed  Google Scholar 

  175. Asano, R. et al. Construction and humanization of a functional bispecific EGFR × CD16 diabody using a refolding system. FEBS J. 279, 223–233 (2012).

    Article  CAS  PubMed  Google Scholar 

  176. Reusch, U. et al. A novel tetravalent bispecific TandAb (CD30/CD16A) efficiently recruits NK cells for the lysis of CD30+ tumor cells. MAbs 6, 727–738 (2014).

    Article  PubMed Central  Google Scholar 

  177. Kellner, C. et al. Heterodimeric bispecific antibody-derivatives against CD19 and CD16 induce effective antibody-dependent cellular cytotoxicity against B-lymphoid tumor cells. Cancer Lett. 303, 128–139 (2011).

    Article  CAS  PubMed  Google Scholar 

  178. Bruenke, J. et al. A recombinant bispecific single-chain Fv antibody against HLA class II and FcγRIII (CD16) triggers effective lysis of lymphoma cells. Br. J. Haematol. 125, 167–179 (2004).

    Article  CAS  PubMed  Google Scholar 

  179. Hartmann, F. et al. Anti-CD16/CD30 bispecific antibody treatment for Hodgkin's disease role of infusion schedule and costimulation with cytokines. Clin. Cancer Res. 7, 1873–1881 (2001).

    CAS  PubMed  Google Scholar 

  180. Kasuya, K. et al. Bispecific anti-HER2 and CD16 single-chain antibody production prolongs the use of stem cell-like cell transplantation against HER2-overexpressing cancer. Int. J. Mol. Med. 25, 209–215 (2010).

    CAS  PubMed  Google Scholar 

  181. Shahied, L. S. et al. Bispecific minibodies targeting HER2/neu and CD16 exhibit improved tumor lysis when placed in a divalent tumor antigen binding format. J. Biol. Chem. 279, 53907–53914 (2004).

    Article  CAS  PubMed  Google Scholar 

  182. Singer, H. et al. Effective elimination of acute myeloid leukemic cells by recombinant bispecific antibody derivatives directed against CD33 and CD16: J. Immunother. 33, 599–608 (2010).

    Article  CAS  PubMed  Google Scholar 

  183. Wiernik, A. et al. Targeting natural killer cells to acute myeloid leukemia in vitro with a CD16 × 33 bispecific killer cell engager and ADAM17 inhibition. Clin. Cancer Res. 19, 3844–3855 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  184. Vallera, D. A. et al. Heterodimeric bispecific single-chain variable-fragment antibodies against EpCAM and CD16 induce effective antibody-dependent cellular cytotoxicity against human carcinoma cells. Cancer Biother. Radiopharm. 28, 274–282 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  185. Spear, P., Wu, M.-R., Sentman, M.-L. & Sentman, C. L. NKG2D ligands as therapeutic targets. Cancer Immun. 13, 8 (2013).

    PubMed  PubMed Central  Google Scholar 

  186. Strandmann, E. P. von et al. A novel bispecific protein (ULBP2-BB4) targeting the NKG2D receptor on natural killer (NK) cells and CD138 activates NK cells and has potent antitumor activity against human multiple myeloma in vitro and in vivo. Blood 107, 1955–1962 (2006).

    Article  CAS  Google Scholar 

  187. Rothe, A. et al. The bispecific immunoligand ULBP2-aCEA redirects natural killer cells to tumor cells and reveals potent anti-tumor activity against colon carcinoma. Int. J. Cancer 134, 2829–2840 (2014).

    Article  CAS  PubMed  Google Scholar 

  188. Germain, C. et al. MHC Class I-related chain A conjugated to antitumor antibodies can sensitize tumor cells to specific lysis by natural killer cells. Clin. Cancer Res. 11, 7516–7522 (2005).

    Article  CAS  PubMed  Google Scholar 

  189. Stamova, S. et al. Simultaneous engagement of the activatory receptors NKG2D and CD3 for retargeting of effector cells to CD33-positive malignant cells. Leukemia 25, 1053–1056 (2011).

    Article  CAS  PubMed  Google Scholar 

Download references

Acknowledgements

L.L.L. is an American Cancer Society professor and is funded by US National Institutes of Health grants AI066897 and AI068129. M.G.M. is a postdoctoral fellow and is funded by the Department of Defense Congressionally Directed Breast Cancer Research Program award W81XWH-13-1-0041.

Author information

Authors and Affiliations

Authors

Corresponding author

Correspondence to Lewis L. Lanier.

Ethics declarations

Competing interests

L.L.L and the University of California San Francisco have licensed intellectual property rights regarding NKG2D for commercial applications. M.G.M. declares no competing interests.

PowerPoint slides

Glossary

Innate lymphoid cell

(ILC). A lymphocyte that participates in the innate immune responses. ILCs are present in RAG-deficient (Rag1−/− and Rag2−/−) mice, so do not require gene rearrangement for their development or recognition.

Natural cytotoxicity receptors

(NCRs). A family of activating receptors expressed by NK cells and some innate lymphoid cells that recognize various, mostly ill-defined, ligands. The three members of this family in humans are NKp46 (encoded by NCR1), NKp44 (encoded by NCR2) and NKp30 (encoded by NCR3). Mice possess only a functional Ncr1 gene and not orthologues of NKp30 or NKp44.

γδT cells

A subset of T cells that express a distinct T cell receptor (TCR) composed of one γ-chain and one δ-chain, instead of the α- and β-chains that comprise the TCR on the majority of T cells. Like natural killer cells, they are considered to be a bridge between innate and adaptive immunity.

Chédiak–Higashi syndrome and X-linked lymphoproliferative syndrome

Autosomal recessive genetic disorders that impair natural killer cell and T cell effector functions.

Invariant natural killer T cells

(iNKT cells). A subset of T cells that express certain natural killer cell markers as well as an invariant T cell receptor α-chain. Their recognition is restricted to glycolipids presented by the CD1d antigen-presenting molecule.

Lymphokine-activated killer cells

(LAK cells). Cytolytic lymphocytes, predominantly natural killer cells, that have been generated after stimulation with interleukin-2 and can spontaneously lyse cancer cells.

Alloantigen-specific response

An immune response directed towards polymorphic antigens expressed by cells of the same species, such as those from transplanted cells or tissues.

Graft-versus-host disease

(GVHD). A complication of allogeneic haematopoietic stem cell transplantation that occurs when contaminating T cells present in the graft recognize the recipient cells as foreign and mount an immune response against them.

Adoptive cell transfer

Refers to the administration of autologous or allogeneic cells to a recipient.

Anergic state

Characterized by the unresponsiveness of immune cells as a result of chronic stimulation or other immunosuppressive mechanisms.

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Morvan, M., Lanier, L. NK cells and cancer: you can teach innate cells new tricks. Nat Rev Cancer 16, 7–19 (2016). https://doi.org/10.1038/nrc.2015.5

Download citation

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/nrc.2015.5

This article is cited by

Search

Quick links

Nature Briefing: Cancer

Sign up for the Nature Briefing: Cancer newsletter — what matters in cancer research, free to your inbox weekly.

Get what matters in cancer research, free to your inbox weekly. Sign up for Nature Briefing: Cancer