Skip to main content

Advertisement

Log in

Current progress in NK cell biology and NK cell-based cancer immunotherapy

  • Focussed Research Review
  • Published:
Cancer Immunology, Immunotherapy Aims and scope Submit manuscript

Abstract

A better understanding of the complex interactions between the immune system and tumour cells from different origins has opened the possibility to design novel procedures of antitumoral immunotherapy. One of these novel approaches is based on the use of autologous or allogeneic natural killer (NK) cells to treat cancer. In the last decade, different strategies to activate NK cells and their use in adoptive NK cell-based therapy have been established. Although NK cells are often considered as a uniform cell population, several phenotypic and functionally distinct NK cells subsets exist in healthy individuals, that are differentially affected by ageing or by apparently innocuous viruses such as cytomegalovirus (CMV). In addition, further alterations in the expression of activating and inhibitory receptors are found in NK cells from cancer patients, likely because of their interaction with tumour cells. Thus, NK cells represent a promising strategy for adoptive immunotherapy of cancer already tested in phase 1/2 clinical trials. However, the existence of NK cell subpopulations expressing different patterns of activating and inhibitory receptors and different functional capacities, that can be found to be altered not only in cancer patients but also in healthy individuals stratified by age or CMV infection, makes necessary a personalized definition of the procedures used in the selection, expansion, and activation of the relevant NK cell subsets to be successfully used in NK cell-based immunotherapy.

This is a preview of subscription content, log in via an institution to check access.

Access this article

Price excludes VAT (USA)
Tax calculation will be finalised during checkout.

Instant access to the full article PDF.

Fig. 1
Fig. 2
Fig. 3

Similar content being viewed by others

Abbreviations

AML:

Acute myeloid leukaemia

ADCC:

Antibody-dependent cell cytotoxicity

Anti-TAA:

Anti-tumour-associated antigens

APCs:

Antigen-presenting cells

CAR:

Chimeric antigen receptor

CMV:

Cytomegalovirus

EGFR:

Epidermal growth factor receptor

GMP:

Good manufacturing practice

GvHD:

Graft-versus-host disease

HCMV:

Human cytomegalovirus

HLA:

Human leukocyte antigen

HSCT:

Hematopoietic stem cell transplantation

KIR:

Killer cell immunoglobulin-like receptors

LAG-3:

Lymphocyte-activating gene 3

MHC:

Major histocompatibility complex

MCMV:

Murine cytomegalovirus

MM:

Multiple myeloma

NCRs:

Natural cytotoxicity receptors

NHL:

Non-Hodgkin lymphoma

NK:

Natural killer

PD-1:

Programmed death-1

TAA:

Tumour-associated antigen

TIGIT:

T cell immunoreceptor with Ig and ITIM domains

TIM-3:

T cell immunoglobulin and mucin domain 3

References

  1. Imai K, Matsuyama S, Miyake S, Suga K, Nakachi K (2000) Natural cytotoxic activity of peripheral-blood lymphocytes and cancer incidence: an 11-year follow-up study of a general population. Lancet 356:1795–1799. https://doi.org/10.1016/S0140-6736(00)03231-1

    Article  CAS  PubMed  Google Scholar 

  2. Ottinger HD, Beelen DW, Scheulen B, Schaefer UW, Grosse-Wilde H (1996) Improved immune reconstitution after allotransplantation of peripheral blood stem cells instead of bone marrow. Blood 88:2775–2779

    CAS  PubMed  Google Scholar 

  3. Zaghi E, Calvi M, Di VC, Mavilio D (2019) Innate immune responses in the outcome of haploidentical hematopoietic stem cell transplantation to cure hematologic malignancies. Front Immunol 10:2794

    PubMed  PubMed Central  Google Scholar 

  4. Ullah MA, Hill GR, Tey SK (2016) Functional reconstitution of natural killer cells in allogeneic hematopoietic stem cell transplantation. Front Immunol 7:144

    PubMed  PubMed Central  Google Scholar 

  5. Minculescu L, Marquart HV, Friis LS, Petersen SL, Schiodt I, Ryder LP, Andersen NS, Sengeloev H (2016) Early natural killer cell reconstitution predicts overall survival in T cell-replete allogeneic hematopoietic stem cell transplantation. Biol Blood Marrow Transplant 22:2187–2193. https://doi.org/10.1016/j.bbmt.2016.09.006

    Article  CAS  PubMed  Google Scholar 

  6. Ruggeri L, Mancusi A, Capanni M, Urbani E, Carotti A, Aloisi T, Stern M, Pende D, Perruccio K, Burchielli E, Topini F, Bianchi E, Aversa F, Martelli MF, Velardi A (2007) Donor natural killer cell allorecognition of missing self in haploidentical hematopoietic transplantation for acute myeloid leukemia: challenging its predictive value. Blood 110:433–440. https://doi.org/10.1182/blood-2006-07-038687

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  7. Hsu KC, Keever-Taylor CA, Wilton A, Pinto C, Heller G, Arkun K, O’Reilly RJ, Horowitz MM, Dupont B (2005) Improved outcome in HLA-identical sibling hematopoietic stem-cell transplantation for acute myelogenous leukemia predicted by KIR and HLA genotypes. Blood 105:4878–4884. https://doi.org/10.1182/blood-2004-12-4825

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  8. Tarazona R, Sanchez-Correa B, Casas-Aviles I, Campos C, Pera A, Morgado S, Lopez-Sejas N, Hassouneh F, Bergua JM, Arcos MJ, Banas H, Casado JG, Duran E, Labella F, Solana R (2017) Immunosenescence: limitations of natural killer cell-based cancer immunotherapy. Cancer Immunol Immunother 66:233–245. https://doi.org/10.1007/s00262-016-1882-x

    Article  CAS  PubMed  Google Scholar 

  9. Sanchez-Correa B, Campos C, Pera A, Bergua JM, Arcos MJ, Banas H, Casado JG, Morgado S, Duran E, Solana R, Tarazona R (2016) Natural killer cell immunosenescence in acute myeloid leukaemia patients: new targets for immunotherapeutic strategies? Cancer Immunol Immunother 65:453–463. https://doi.org/10.1007/s00262-015-1720-6

    Article  CAS  PubMed  Google Scholar 

  10. Lichtenegger FS, Krupka C, Kohnke T, Subklewe M (2015) Immunotherapy for acute myeloid leukemia. Semin Hematol 52:207–214. https://doi.org/10.1053/j.seminhematol.2015.03.006

    Article  CAS  PubMed  Google Scholar 

  11. Servais S, Porcher R, Xhaard A, Robin M, Masson E, Larghero J, Ribaud P, Dhedin N, Abbes S, Sicre F, Socie G, de Peffault LR (2014) Pre-transplant prognostic factors of long-term survival after allogeneic peripheral blood stem cell transplantation with matched related/unrelated donors. Haematologica 99:519–526

    PubMed  PubMed Central  Google Scholar 

  12. Rezvani AR, Storer BE, Guthrie KA, Schoch HG, Maloney DG, Sandmaier BM, Storb R (2015) Impact of donor age on outcome after allogeneic hematopoietic cell transplantation. Biol Blood Marrow Transplant 21:105–112

    PubMed  Google Scholar 

  13. Parkhurst MR, Riley JP, Dudley ME, Rosenberg SA (2011) Adoptive transfer of autologous natural killer cells leads to high levels of circulating natural killer cells but does not mediate tumor regression. Clin Cancer Res 17:6287–6297. https://doi.org/10.1158/1078-0432.CCR-11-1347

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  14. Veluchamy JP, Kok N, van der Vlet HJ, Verheul HMW, de Gruijl TD, Spanholtz J (2017) The rise of allogeneic natural killer cells as a platform for cancer immunotherapy: recent innovations and future developments. Front Immunol 8:631. https://doi.org/10.3389/fimmu.2017.00631

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  15. Lupo KB, Matosevic S (2019) Natural killer cells as allogeneic effectors in adoptive cancer immunotherapy. Cancers (Basel). https://doi.org/10.3390/cancers11060769

    Article  Google Scholar 

  16. Del Zotto G, Marcenaro E, Vacca P, Sivori S, Pende D, Della CM, Moretta F, Ingegnere T, Mingari MC, Moretta A, Moretta L (2017) Markers and function of human NK cells in normal and pathological conditions. Cytometry B Clin Cytom 92:100–114. https://doi.org/10.1002/cyto.b.21508

    Article  CAS  PubMed  Google Scholar 

  17. Freud AG, Mundy-Bosse BL, Yu J, Caligiuri MA (2017) The broad spectrum of human natural killer cell diversity. Immunity 47:820–833. https://doi.org/10.1016/j.immuni.2017.10.008

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  18. Horowitz A, Strauss-Albee DM, Leipold M, Kubo J, Nemat-Gorgani N, Dogan OC, Dekker CL, Mackey S, Maecker H, Swan GE, Davis MM, Norman PJ, Guethlein LA, Desai M, Parham P, Blish CA (2013) Genetic and environmental determinants of human NK cell diversity revealed by mass cytometry. Sci Transl Med 5:208ra145. https://doi.org/10.1126/scitranslmed.3006702

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  19. Di Vito C, Mikulak J, Mavilio D (2019) On the way to become a natural killer cell. Front Immunol 10:1812. https://doi.org/10.3389/fimmu.2019.01812

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  20. Morgado S, Sanchez-Correa B, Casado JG, Duran E, Gayoso I, Labella F, Solana R, Tarazona R (2011) NK cell recognition and killing of melanoma cells is controlled by multiple activating receptor-ligand interactions. J Innate Immun 3:365–373. https://doi.org/10.1159/000328505

    Article  CAS  PubMed  Google Scholar 

  21. Paul S, Lal G (2017) The molecular mechanism of natural killer cells function and its importance in cancer immunotherapy. Front Immunol 8:1124. https://doi.org/10.3389/fimmu.2017.01124

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  22. Vivier E, Raulet DH, Moretta A, Caligiuri MA, Zitvogel L, Lanier LL, Yokoyama WM, Ugolini S (2011) Innate or adaptive immunity? The example of natural killer cells. Science 331:44–49. https://doi.org/10.1126/science.1198687

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  23. Sanchez-Correa B, Lopez-Sejas N, Duran E, Labella F, Alonso C, Solana R, Tarazona R (2019) Modulation of NK cells with checkpoint inhibitors in the context of cancer immunotherapy. Cancer Immunol Immunother 68:861–870. https://doi.org/10.1007/s00262-019-02336-6

    Article  CAS  PubMed  Google Scholar 

  24. Sanchez-Correa B, Valhondo I, Hassouneh F, Lopez-Sejas N, Pera A, Bergua JM, Arcos MJ, Banas H, Casas-Aviles I, Duran E, Alonso C, Solana R, Tarazona R (2019) DNAM-1 and the TIGIT/PVRIG/TACTILE axis: novel immune checkpoints for natural killer cell-based cancer immunotherapy. Cancers (Basel). https://doi.org/10.3390/cancers11060877

    Article  Google Scholar 

  25. Anderson AC, Joller N, Kuchroo VK (2016) Lag-3, Tim-3, and TIGIT: co-inhibitory receptors with specialized functions in immune regulation. Immunity 44:989–1004. https://doi.org/10.1016/j.immuni.2016.05.001

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  26. Joller N, Kuchroo VK (2017) Tim-3, Lag-3, and TIGIT. Curr Top Microbiol Immunol 410:127–156. https://doi.org/10.1007/82_2017_62

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  27. Bauer S, Groh V, Wu J, Steinle A, Phillips JH, Lanier LL, Spies T (1999) Activation of NK cells and T cells by NKG2D, a receptor for stress-inducible MICA. Science 285:727–729. https://doi.org/10.1126/science.285.5428.727

    Article  CAS  PubMed  Google Scholar 

  28. Sivori S, Vitale M, Morelli L, Sanseverino L, Augugliaro R, Bottino C, Moretta L, Moretta A (1997) p46, a novel natural killer cell-specific surface molecule that mediates cell activation. J Exp Med 186:1129–1136. https://doi.org/10.1084/jem.186.7.1129

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  29. Pende D, Parolini S, Pessino A, Sivori S, Augugliaro R, Morelli L, Marcenaro E, Accame L, Malaspina A, Biassoni R, Bottino C, Moretta L, Moretta A (1999) 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. https://doi.org/10.1084/jem.190.10.1505

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  30. Vitale M, Bottino C, Sivori S, Sanseverino L, Castriconi R, Marcenaro E, Augugliaro R, Moretta L, Moretta A (1998) NKp44, a novel triggering surface molecule specifically expressed by activated natural killer cells, is involved in non-major histocompatibility complex-restricted tumor cell lysis. J Exp Med 187:2065–2072. https://doi.org/10.1084/jem.187.12.2065

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  31. Shibuya A, Campbell D, Hannum C, Yssel H, Franz-Bacon K, McClanahan T, Kitamura T, Nicholl J, Sutherland GR, Lanier LL, Phillips JH (1996) DNAM-1, a novel adhesion molecule involved in the cytolytic function of T lymphocytes. Immunity 4:573–581. https://doi.org/10.1016/S1074-7613(00)70060-4

    Article  CAS  PubMed  Google Scholar 

  32. Casado JG, Pawelec G, Morgado S, Sanchez-Correa B, Delgado E, Gayoso I, Duran E, Solana R, Tarazona R (2009) Expression of adhesion molecules and ligands for activating and costimulatory receptors involved in cell-mediated cytotoxicity in a large panel of human melanoma cell lines. Cancer Immunol Immunother 58:1517–1526. https://doi.org/10.1007/s00262-009-0682-y

    Article  CAS  PubMed  Google Scholar 

  33. Sanchez-Correa B, Morgado S, Gayoso I, Bergua JM, Casado JG, Arcos MJ, Bengochea ML, Duran E, Solana R, Tarazona R (2011) Human NK cells in acute myeloid leukaemia patients: analysis of NK cell-activating receptors and their ligands. Cancer Immunol Immunother 60:1195–1205. https://doi.org/10.1007/s00262-011-1050-2

    Article  CAS  PubMed  Google Scholar 

  34. Solana R, Campos C, Pera A, Tarazona R (2014) Shaping of NK cell subsets by aging. Curr Opin Immunol 29:56–61. https://doi.org/10.1016/j.coi.2014.04.002

    Article  CAS  PubMed  Google Scholar 

  35. Campos C, Pera A, Sanchez-Correa B, Alonso C, Lopez-Fernandez I, Morgado S, Tarazona R, Solana R (2014) Effect of age and CMV on NK cell subpopulations. Exp Gerontol 54:130–137. https://doi.org/10.1016/j.exger.2014.01.008

    Article  CAS  PubMed  Google Scholar 

  36. Chidrawar SM, Khan N, Chan YL, Nayak L, Moss PA (2006) Ageing is associated with a decline in peripheral blood CD56bright NK cells. Immun Ageing 3:10. https://doi.org/10.1186/1742-4933-3-10

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  37. Fali T, Papagno L, Bayard C, Mouloud Y, Boddaert J, Sauce D, Appay V (2019) New insights into lymphocyte differentiation and aging from telomere length and telomerase activity measurements. J Immunol 202:1962–1969. https://doi.org/10.4049/jimmunol.1801475

    Article  CAS  PubMed  Google Scholar 

  38. Lopez-Verges S, Milush JM, Schwartz BS, Pando MJ, Jarjoura J, York VA, Houchins JP, Miller S, Kang SM, Norris PJ, Nixon DF, Lanier LL (2011) Expansion of a unique CD57(+)NKG2Chi natural killer cell subset during acute human cytomegalovirus infection. Proc Natl Acad Sci USA 108:14725–14732. https://doi.org/10.1073/pnas.1110900108

    Article  PubMed  PubMed Central  Google Scholar 

  39. Bjorkstrom NK, Riese P, Heuts F, Andersson S, Fauriat C, Ivarsson MA, Bjorklund AT, Flodstrom-Tullberg M, Michaelsson J, Rottenberg ME, Guzman CA, Ljunggren HG, Malmberg KJ (2010) Expression patterns of NKG2A, KIR, and CD57 define a process of CD56dim NK cell differentiation uncoupled from NK cell education. Blood 116:3853–3864. https://doi.org/10.1182/blood-2010-04-281675

    Article  CAS  PubMed  Google Scholar 

  40. Campos C, Lopez N, Pera A, Gordillo JJ, Hassouneh F, Tarazona R, Solana R (2015) Expression of NKp30, NKp46 and DNAM-1 activating receptors on resting and IL-2 activated NK cells from healthy donors according to CMV-serostatus and age. Biogerontology 16:671–683. https://doi.org/10.1007/s10522-015-9581-0

    Article  CAS  PubMed  Google Scholar 

  41. Foley B, Cooley S, Verneris MR, Pitt M, Curtsinger J, Luo X, Lopez-Verges S, Lanier LL, Weisdorf D, Miller JS (2012) Cytomegalovirus reactivation after allogeneic transplantation promotes a lasting increase in educated NKG2C+ natural killer cells with potent function. Blood 119:2665–2674. https://doi.org/10.1182/blood-2011-10-386995

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  42. Solana R, Tarazona R, Aiello AE, Akbar AN, Appay V, Beswick M, Bosch JA, Campos C, Cantisan S, Cicin-Sain L, Derhovanessian E, Ferrando-Martinez S, Frasca D, Fulop T, Govind S, Grubeck-Loebenstein B, Hill A, Hurme M, Kern F, Larbi A, Lopez-Botet M, Maier AB, McElhaney JE, Moss P, Naumova E, Nikolich-Zugich J, Pera A, Rector JL, Riddell N, Sanchez-Correa B, Sansoni P, Sauce D, van Rene L, Wang GC, Wills MR, Zielinski M, Pawelec G (2012) CMV and immunosenescence: from basics to clinics. Immun Ageing 9:23. https://doi.org/10.1182/blood-2011-10-386995

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  43. Lopez-Sejas N, Campos C, Hassouneh F, Sanchez-Correa B, Tarazona R, Pera A, Solana R (2016) Effect of CMV and aging on the differential expression of CD300a, CD161, T-bet, and Eomes on NK Cell subsets. Front Immunol 7:476. https://doi.org/10.3389/fimmu.2016.00476

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  44. Hassouneh F, Lopez-Sejas N, Campos C, Sanchez-Correa B, Tarazona R, Solana R, Pera A (2017) Differential effect of cytomegalovirus infection with age on the expression of CD57, CD300a, and CD161 on T-cell subpopulations. Front Immunol 8:649. https://doi.org/10.3389/fimmu.2017.00649

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  45. Pera A, Vasudev A, Tan C, Kared H, Solana R, Larbi A (2017) CMV induces expansion of highly polyfunctional CD4+ T cell subset coexpressing CD57 and CD154. J Leukoc Biol 101:555–566. https://doi.org/10.1189/jlb.4A0316-112R

    Article  CAS  PubMed  Google Scholar 

  46. Pera A, Campos C, Corona A, Sanchez-Correa B, Tarazona R, Larbi A, Solana R (2014) CMV latent infection improves CD8+ T response to SEB due to expansion of polyfunctional CD57+ cells in young individuals. PLoS ONE 9:e88538. https://doi.org/10.1371/journal.pone.0088538

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  47. Lanier LL (2005) NK cell recognition. Annu Rev Immunol 23:225–274

    CAS  PubMed  Google Scholar 

  48. Chiossone L, Chaix J, Fuseri N, Roth C, Vivier E, Walzer T (2009) Maturation of mouse NK cells is a 4-stage developmental program. Blood 113:5488–5496

    CAS  PubMed  Google Scholar 

  49. Loh J, Chu DT, O’Guin AK, Yokoyama WM, Virgin HW (2005) Natural killer cells utilize both perforin and gamma interferon to regulate murine cytomegalovirus infection in the spleen and liver. J Virol 79:661–667

    CAS  PubMed  PubMed Central  Google Scholar 

  50. Fang M, Lanier LL, Sigal LJ (2008) A role for NKG2D in NK cell-mediated resistance to poxvirus disease. PLoS Pathog 4:e30

    PubMed  PubMed Central  Google Scholar 

  51. Beli E, Clinthorne JF, Duriancik DM, Hwang I, Kim S, Gardner EM (2011) Natural killer cell function is altered during the primary response of aged mice to influenza infection. Mech Ageing Dev 132:503–510

    CAS  PubMed  PubMed Central  Google Scholar 

  52. Fang M, Roscoe F, Sigal LJ (2010) Age-dependent susceptibility to a viral disease due to decreased natural killer cell numbers and trafficking. J Exp Med 207:2369–2381

    CAS  PubMed  PubMed Central  Google Scholar 

  53. Nogusa S, Ritz BW, Kassim SH, Jennings SR, Gardner EM (2008) Characterization of age-related changes in natural killer cells during primary influenza infection in mice. Mech Ageing Dev 129:223–230

    CAS  PubMed  Google Scholar 

  54. Duan X, Lu J, Wang H, Liu X, Wang J, Zhou K, Jiang W, Wang Y, Fang M (2017) Bidirectional factors impact the migration of NK cells to draining lymph node in aged mice during influenza virus infection. Exp Gerontol 96:127–137

    CAS  PubMed  Google Scholar 

  55. Beli E, Duriancik DM, Clinthorne JF, Lee T, Kim S, Gardner EM (2014) Natural killer cell development and maturation in aged mice. Mech Ageing Dev 135:33–40

    CAS  PubMed  Google Scholar 

  56. Shehata HM, Hoebe K, Chougnet CA (2015) The aged nonhematopoietic environment impairs natural killer cell maturation and function. Aging Cell 14:191–199

    CAS  PubMed  PubMed Central  Google Scholar 

  57. Chiu BC, Martin BE, Stolberg VR, Chensue SW (2013) The host environment is responsible for aging-related functional NK cell deficiency. J Immunol 191:4688–4698

    CAS  PubMed  Google Scholar 

  58. Nair S, Fang M, Sigal LJ (2015) The natural killer cell dysfunction of aged mice is due to the bone marrow stroma and is not restored by IL-15/IL-15Ralpha treatment. Aging Cell 14:180–190

    CAS  PubMed  Google Scholar 

  59. Smith HR, Heusel JW, Mehta IK, Kim S, Dorner BG, Naidenko OV, Iizuka K, Furukawa H, Beckman DL, Pingel JT, Scalzo AA, Fremont DH, Yokoyama WM (2002) Recognition of a virus-encoded ligand by a natural killer cell activation receptor. Proc Natl Acad Sci USA 99:8826–8831. https://doi.org/10.1073/pnas.092258599

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  60. Arase H, Mocarski ES, Campbell AE, Hill AB, Lanier LL (2002) Direct recognition of cytomegalovirus by activating and inhibitory NK cell receptors. Science 296:1323–1326. https://doi.org/10.1126/science.1070884

    Article  CAS  PubMed  Google Scholar 

  61. Orr MT, Sun JC, Hesslein DG, Arase H, Phillips JH, Takai T, Lanier LL (2009) Ly49H signaling through DAP10 is essential for optimal natural killer cell responses to mouse cytomegalovirus infection. J Exp Med 206:807–817

    CAS  PubMed  PubMed Central  Google Scholar 

  62. Sun JC, Madera S, Bezman NA, Beilke JN, Kaplan MH, Lanier LL (2012) Proinflammatory cytokine signaling required for the generation of natural killer cell memory. J Exp Med 209:947–954. https://doi.org/10.1084/jem.20111760

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  63. Guma M, Budt M, Saez A, Brckalo T, Hengel H, Angulo A, Lopez-Botet M (2006) Expansion of CD94/NKG2C+ NK cells in response to human cytomegalovirus-infected fibroblasts. Blood 107:3624–3631

    CAS  PubMed  Google Scholar 

  64. Lopez-Botet M, Muntasell A, Vilches C (2014) The CD94/NKG2C+ NK-cell subset on the edge of innate and adaptive immunity to human cytomegalovirus infection. Semin Immunol 26:145–151

    CAS  PubMed  Google Scholar 

  65. Muntasell A, Vilches C, Angulo A, Lopez-Botet M (2013) Adaptive reconfiguration of the human NK-cell compartment in response to cytomegalovirus: a different perspective of the host-pathogen interaction. Eur J Immunol 43:1133–1141

    CAS  PubMed  Google Scholar 

  66. Bjorkstrom NK, Lindgren T, Stoltz M, Fauriat C, Braun M, Evander M, Michaelsson J, Malmberg KJ, Klingstrom J, Ahlm C, Ljunggren HG (2011) Rapid expansion and long-term persistence of elevated NK cell numbers in humans infected with hantavirus. J Exp Med 208:13–21

    PubMed  PubMed Central  Google Scholar 

  67. Petitdemange C, Becquart P, Wauquier N, Beziat V, Debre P, Leroy EM, Vieillard V (2011) Unconventional repertoire profile is imprinted during acute chikungunya infection for natural killer cells polarization toward cytotoxicity. PLoS Pathog 7:e1002268

    CAS  PubMed  PubMed Central  Google Scholar 

  68. Hendricks DW, Balfour HH Jr, Dunmire SK, Schmeling DO, Hogquist KA, Lanier LL (2014) Cutting edge: NKG2C(hi)CD57+ NK cells respond specifically to acute infection with cytomegalovirus and not Epstein-Barr virus. J Immunol 192:4492–4496

    CAS  PubMed  Google Scholar 

  69. Merino A, Zhang B, Dougherty P, Luo X, Wang J, Blazar BR, Miller JS, Cichocki F (2019) Chronic stimulation drives human NK cell dysfunction and epigenetic reprograming. J Clin Invest 130:3770–3785

    Google Scholar 

  70. Paust S, Gill HS, Wang BZ, Flynn MP, Moseman EA, Senman B, Szczepanik M, Telenti A, Askenase PW, Compans RW, von Andrian UH (2010) Critical role for the chemokine receptor CXCR6 in NK cell-mediated antigen-specific memory of haptens and viruses. Nat Immunol 11:1127–1135

    CAS  PubMed  PubMed Central  Google Scholar 

  71. Nikzad R, Angelo LS, Aviles-Padilla K, Le DT, Singh VK, Bimler L, Vukmanovic-Stejic M, Vendrame E, Ranganath T, Simpson L, Haigwood NL, Blish CA, Akbar AN, Paust S (2019) Human natural killer cells mediate adaptive immunity to viral antigens. Sci Immunol 4:eaat8116

    CAS  PubMed  PubMed Central  Google Scholar 

  72. Hydes T, Abuhilal M, Armstrong T, Primrose J, Takhar A, Khakoo S (2015) Natural killer cell maturation markers in the human liver and expansion of an NKG2C+ KIR+ population. Lancet 385(Suppl 1):S45

    PubMed  Google Scholar 

  73. Sivori S, Meazza R, Quintarelli C, Carlomagno S, Della CM, Falco M, Moretta L, Locatelli F, Pende D (2019) NK Cell-based immunotherapy for hematological malignancies. J Clin Med. https://doi.org/10.3390/jcm8101702

    Article  PubMed  PubMed Central  Google Scholar 

  74. Verheyden S, Bernier M, Demanet C (2004) Identification of natural killer cell receptor phenotypes associated with leukemia. Leukemia 18:2002–2007. https://doi.org/10.1038/sj.leu.2403525

    Article  CAS  PubMed  Google Scholar 

  75. Zhang Y, Wang B, Ye S, Liu S, Liu M, Shen C, Teng Y, Qi J (2010) Killer cell immunoglobulin-like receptor gene polymorphisms in patients with leukemia: possible association with susceptibility to the disease. Leuk Res 34:55–58. https://doi.org/10.1016/j.leukres.2009.04.022

    Article  CAS  PubMed  Google Scholar 

  76. He Y, Bunn PA, Zhou C, Chan D (2016) KIR 2D (L1, L3, L4, S4) and KIR 3DL1 protein expression in non-small cell lung cancer. Oncotarget 7:82104–82111. https://doi.org/10.18632/oncotarget.13486

    Article  PubMed  PubMed Central  Google Scholar 

  77. Fauriat C, Just-Landi S, Mallet F, Arnoulet C, Sainty D, Olive D, Costello RT (2007) 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 109:323–330. https://doi.org/10.1182/blood-2005-08-027979

    Article  CAS  PubMed  Google Scholar 

  78. Stringaris K, Sekine T, Khoder A, Alsuliman A, Razzaghi B, Sargeant R, Pavlu J, Brisley G, de Lavallade H, Sarvaria A, Marin D, Mielke S, Apperley JF, Shpall EJ, Barrett AJ, Rezvani K (2014) Leukemia-induced phenotypic and functional defects in natural killer cells predict failure to achieve remission in acute myeloid leukemia. Haematologica 99:836–847. https://doi.org/10.3324/haematol.2013.087536

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  79. Pasero C, Gravis G, Granjeaud S, Guerin M, Thomassin-Piana J, Rocchi P, Salem N, Walz J, Moretta A, Olive D (2015) Highly effective NK cells are associated with good prognosis in patients with metastatic prostate cancer. Oncotarget 6:14360–14373. https://doi.org/10.18632/oncotarget.3965

    Article  PubMed  PubMed Central  Google Scholar 

  80. Sun C, Xu J, Huang Q, Huang M, Wen H, Zhang C, Wang J, Song J, Zheng M, Sun H, Wei H, Xiao W, Sun R, Tian Z (2017) High NKG2A expression contributes to NK cell exhaustion and predicts a poor prognosis of patients with liver cancer. Oncoimmunology 6:e1264562. https://doi.org/10.1080/2162402X.2016.1264562

    Article  CAS  PubMed  Google Scholar 

  81. Sanchez-Correa B, Gayoso I, Bergua JM, Casado JG, Morgado S, Solana R, Tarazona R (2012) Decreased expression of DNAM-1 on NK cells from acute myeloid leukemia patients. Immunol Cell Biol 90:109–115. https://doi.org/10.1038/icb.2011.15

    Article  CAS  PubMed  Google Scholar 

  82. Sanchez-Correa B, Bergua JM, Pera A, Campos C, Arcos MJ, Banas H, Duran E, Solana R, Tarazona R (2017) In vitro culture with interleukin-15 leads to expression of activating receptors and recovery of natural killer cell function in acute myeloid leukemia patients. Front Immunol 8:931. https://doi.org/10.3389/fimmu.2017.00931

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  83. Almeida-Oliveira A, Smith-Carvalho M, Porto LC, Cardoso-Oliveira J, Ribeiro AS, Falcao RR, Abdelhay E, Bouzas LF, Thuler LC, Ornellas MH, Diamond HR (2011) Age-related changes in natural killer cell receptors from childhood through old age. Hum Immunol 72:319–329. https://doi.org/10.1016/j.humimm.2011.01.009

    Article  CAS  PubMed  Google Scholar 

  84. Liu H, Wang S, Xin J, Wang J, Yao C, Zhang Z (2019) Role of NKG2D and its ligands in cancer immunotherapy. Am J Cancer Res 9:2064–2078

    PubMed  PubMed Central  Google Scholar 

  85. Kaifu T, Nakamura A (2017) Polymorphisms of immunoglobulin receptors and the effects on clinical outcome in cancer immunotherapy and other immune diseases: a general review. Int Immunol 29:319–325. https://doi.org/10.1093/intimm/dxx041

    Article  CAS  PubMed  Google Scholar 

  86. Tarazona R, Casado JG, Soto R, DelaRosa O, Peralbo E, Rioja L, Pena J, Solana R (2004) Expression of NK-associated receptors on cytotoxic T cells from melanoma patients: a two-edged sword? Cancer Immunol Immunother 53:911–924

    CAS  PubMed  Google Scholar 

  87. Hofer E, Koehl U (2017) Natural killer cell-based cancer immunotherapies: from immune evasion to promising targeted cellular therapies. Front Immunol 8:745

    PubMed  PubMed Central  Google Scholar 

  88. Rosenberg SA, Lotze MT, Muul LM, Leitman S, Chang AE, Ettinghausen SE, Matory YL, Skibber JM, Shiloni E, Vetto JT (1985) 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. https://doi.org/10.1056/NEJM198512053132327

    Article  CAS  PubMed  Google Scholar 

  89. Pangrazzi L, Meryk A, Naismith E, Koziel R, Lair J, Krismer M, Trieb K, Grubeck-Loebenstein B (2017) “Inflamm-aging” influences immune cell survival factors in human bone marrow. Eur J Immunol 47:481–492

    CAS  PubMed  PubMed Central  Google Scholar 

  90. Yalcin A, Silay K, Balik AR, Avcioglu G, Aydin AS (2018) The relationship between plasma interleukin-15 levels and sarcopenia in outpatient older people. Aging Clin Exp Res 30:783–790

    PubMed  Google Scholar 

  91. Al-Attar A, Presnell SR, Clasey JL, Long DE, Walton RG, Sexton M, Starr ME, Kern PA, Peterson CA, Lutz CT (2018) Human body composition and immunity: visceral adipose tissue produces IL-15 and muscle strength inversely correlates with NK cell function in elderly humans. Front Immunol 9:440

    PubMed  PubMed Central  Google Scholar 

  92. Ochoa MC, Minute L, Rodriguez I, Garasa S, Perez-Ruiz E, Inoges S, Melero I, Berraondo P (2017) Antibody-dependent cell cytotoxicity: immunotherapy strategies enhancing effector NK cells. Immunol Cell Biol 95:347–355

    CAS  PubMed  Google Scholar 

  93. Gluck WL, Hurst D, Yuen A, Levine AM, Dayton MA, Gockerman JP, Lucas J, Denis-Mize K, Tong B, Navis D, Difrancesco A, Milan S, Wilson SE, Wolin M (2004) Phase I studies of interleukin (IL)-2 and rituximab in B-cell non-Hodgkin’s lymphoma: IL-2 mediated natural killer cell expansion correlations with clinical response. Clin Cancer Res 10:2253–2264. https://doi.org/10.1158/1078-0432.CCR-1087-3

    Article  CAS  PubMed  Google Scholar 

  94. Leivas A, Perez-Martinez A, Blanchard MJ, Martin-Clavero E, Fernandez L, Lahuerta JJ, Martinez-Lopez J (2016) Novel treatment strategy with autologous activated and expanded natural killer cells plus anti-myeloma drugs for multiple myeloma. Oncoimmunology 5:e1250051. https://doi.org/10.1080/2162402X.2016.1250051

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  95. Rubnitz JE, Inaba H, Ribeiro RC, Pounds S, Rooney B, Bell T, Pui CH, Leung W (2010) NKAML: a pilot study to determine the safety and feasibility of haploidentical natural killer cell transplantation in childhood acute myeloid leukemia. J Clin Oncol 28:955–959. https://doi.org/10.1200/JCO.2009.24.4590

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  96. Ansell SM, Geyer SM, Maurer MJ, Kurtin PJ, Micallef IN, Stella P, Etzell P, Novak AJ, Erlichman C, Witzig TE (2006) Randomized phase II study of interleukin-12 in combination with rituximab in previously treated non-Hodgkin’s lymphoma patients. Clin Cancer Res 12:6056–6063. https://doi.org/10.1158/1078-0432.CCR-06-1245

    Article  CAS  PubMed  Google Scholar 

  97. Parihar R, Nadella P, Lewis A, Jensen R, De HC, Dierksheide JE, VanBuskirk AM, Magro CM, Young DC, Shapiro CL, Carson WE III (2004) A phase I study of interleukin 12 with trastuzumab in patients with human epidermal growth factor receptor-2-overexpressing malignancies: analysis of sustained interferon gamma production in a subset of patients. Clin Cancer Res 10:5027–5037. https://doi.org/10.1158/1078-0432.CCR-04-0265

    Article  CAS  PubMed  Google Scholar 

  98. Charych DH, Hoch U, Langowski JL, Lee SR, Addepalli MK, Kirk PB, Sheng D, Liu X, Sims PW, VanderVeen LA, Ali CF, Chang TK, Konakova M, Pena RL, Kanhere RS, Kirksey YM, Ji C, Wang Y, Huang J, Sweeney TD, Kantak SS, Doberstein SK (2016) NKTR-214, an engineered cytokine with biased IL2 receptor binding, increased tumor exposure, and marked efficacy in mouse tumor models. Clin Cancer Res 22:680–690

    CAS  PubMed  Google Scholar 

  99. Bentebibel SE, Hurwitz ME, Bernatchez C, Haymaker C, Hudgens CW, Kluger HM, Tetzlaff MT, Tagliaferri MA, Zalevsky J, Hoch U, Fanton C, Aung S, Hwu P, Curti BD, Tannir NM, Sznol M, Diab A (2019) A first-in-human study and biomarker analysis of NKTR-214, a novel IL2Rbetagamma-biased cytokine, in patients with advanced or metastatic solid tumors. Cancer Discov 9:711–721

    PubMed  Google Scholar 

  100. Croce M, Rigo V, Ferrini S (2015) IL-21: a pleiotropic cytokine with potential applications in oncology. J Immunol Res 2015:696578. https://doi.org/10.1155/2015/696578

    Article  PubMed  PubMed Central  Google Scholar 

  101. Wu Y, Tian Z, Wei H (2017) Developmental and functional control of natural killer cells by cytokines. Front Immunol 8:930. https://doi.org/10.3389/fimmu.2017.00930

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  102. Gleason MK, Ross JA, Warlick ED, Lund TC, Verneris MR, Wiernik A, Spellman S, Haagenson MD, Lenvik AJ, Litzow MR, Epling-Burnette PK, Blazar BR, Weiner LM, Weisdorf DJ, Vallera DA, Miller JS (2014) CD16xCD33 bispecific killer cell engager (BiKE) activates NK cells against primary MDS and MDSC CD33+ targets. Blood 123:3016–3026

    CAS  PubMed  PubMed Central  Google Scholar 

  103. Davis ZB, Vallera DA, Miller JS, Felices M (2017) Natural killer cells unleashed: checkpoint receptor blockade and BiKE/TriKE utilization in NK-mediated anti-tumor immunotherapy. Semin Immunol 31:64–75. https://doi.org/10.1016/j.smim.2017.07.011

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  104. Wang LX, Tong X, Li C, Giddens JP, Li T (2019) Glycoengineering of antibodies for modulating functions. Annu Rev Biochem 88:433–459. https://doi.org/10.1146/annurev-biochem-062917-012911

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  105. Kim N, Kim HS (2018) Targeting checkpoint receptors and molecules for therapeutic modulation of natural killer cells. Front Immunol 9(2041):2041. https://doi.org/10.3389/fimmu.2018.02041

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  106. Pesce S, Greppi M, Grossi F, Del ZG, Moretta L, Sivori S, Genova C, Marcenaro E (2019) PD/1-PD-Ls checkpoint: insight on the potential role of NK cells. Front Immunol 10:1242. https://doi.org/10.3389/fimmu.2019.01242

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  107. Andre P, Denis C, Soulas C, Bourbon-Caillet C, Lopez J, Arnoux T, Blery M, Bonnafous C, Gauthier L, Morel A, Rossi B, Remark R, Breso V, Bonnet E, Habif G, Guia S, Lalanne AI, Hoffmann C, Lantz O, Fayette J, Boyer-Chammard A, Zerbib R, Dodion P, Ghadially H, Jure-Kunkel M, Morel Y, Herbst R, Narni-Mancinelli E, Cohen RB, Vivier E (2018) Anti-NKG2A mAb is a checkpoint inhibitor that promotes anti-tumor immunity by unleashing both T and NK cells. Cell 175:1731–1743. https://doi.org/10.1016/j.cell.2018.10.014

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  108. Vey N, Karlin L, Sadot-Lebouvier S, Broussais F, Berton-Rigaud D, Rey J, Charbonnier A, Marie D, Andre P, Paturel C, Zerbib R, Bennouna J, Salles G, Goncalves A (2018) A phase 1 study of lirilumab (antibody against killer immunoglobulin-like receptor antibody KIR2D; IPH2102) in patients with solid tumors and hematologic malignancies. Oncotarget 9:17675–17688. https://doi.org/10.18632/oncotarget.24832

    Article  PubMed  PubMed Central  Google Scholar 

  109. Romagne F, Andre P, Spee P, Zahn S, Anfossi N, Gauthier L, Capanni M, Ruggeri L, Benson DM Jr, Blaser BW, Della CM, Moretta A, Vivier E, Caligiuri MA, Velardi A, Wagtmann N (2009) 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

    CAS  PubMed  PubMed Central  Google Scholar 

  110. Carlsten M, Korde N, Kotecha R, Reger R, Bor S, Kazandjian D, Landgren O, Childs RW (2016) Checkpoint Inhibition of KIR2D with the monoclonal antibody IPH2101 induces contraction and hyporesponsiveness of NK cells in patients with myeloma. Clin Cancer Res 22:5211–5222. https://doi.org/10.1158/1078-0432.CCR-16-1108

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  111. Felices M, Miller JS (2016) Targeting KIR blockade in multiple myeloma: trouble in checkpoint paradise? Clin Cancer Res 22:5161–5163. https://doi.org/10.1158/1078-0432.CCR-16-1582

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  112. da Silva I, Gallois A, Jimenez-Baranda S, Khan S, Anderson AC, Kuchroo VK, Osman I, Bhardwaj N (2014) Reversal of NK-cell exhaustion in advanced melanoma by Tim-3 blockade. Cancer Immunol Res 2:410–422. https://doi.org/10.1158/2326-6066.CIR-13-0171

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  113. Xu L, Huang Y, Tan L, Yu W, Chen D, Lu C, He J, Wu G, Liu X, Zhang Y (2015) Increased Tim-3 expression in peripheral NK cells predicts a poorer prognosis and Tim-3 blockade improves NK cell-mediated cytotoxicity in human lung adenocarcinoma. Int Immunopharmacol 29:635–641. https://doi.org/10.1016/j.intimp.2015.09.017

    Article  CAS  PubMed  Google Scholar 

  114. Stanietsky N, Simic H, Arapovic J, Toporik A, Levy O, Novik A, Levine Z, Beiman M, Dassa L, Achdout H, Stern-Ginossar N, Tsukerman P, Jonjic S, Mandelboim O (2009) The interaction of TIGIT with PVR and PVRL2 inhibits human NK cell cytotoxicity. Proc Natl Acad Sci USA 106:17858–17863. https://doi.org/10.1073/pnas.0903474106

    Article  PubMed  PubMed Central  Google Scholar 

  115. Xu F, Sunderland A, Zhou Y, Schulick RD, Edil BH, Zhu Y (2017) Blockade of CD112R and TIGIT signaling sensitizes human natural killer cell functions. Cancer Immunol Immunother 66:1367–1375. https://doi.org/10.1007/s00262-017-2031-x

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  116. Suzuki H, Duncan GS, Takimoto H, Mak TW (1997) Abnormal development of intestinal intraepithelial lymphocytes and peripheral natural killer cells in mice lacking the IL-2 receptor beta chain. J Exp Med 185:499–505. https://doi.org/10.1084/jem.185.3.499

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  117. Choucair K, Duff JR, Cassidy CS, Albrethsen MT, Kelso JD, Lenhard A, Staats H, Patel R, Brunicardi FC, Dworkin L, Nemunaitis J (2019) Natural killer cells: a review of biology, therapeutic potential and challenges in treatment of solid tumors. Future Oncol 15:3053–3069. https://doi.org/10.2217/fon-2019-0116

    Article  CAS  PubMed  Google Scholar 

  118. McKenna DH Jr, Sumstad D, Bostrom N, Kadidlo DM, Fautsch S, McNearney S, Dewaard R, McGlave PB, Weisdorf DJ, Wagner JE, McCullough J, Miller JS (2007) Good manufacturing practices production of natural killer cells for immunotherapy: a six-year single-institution experience. Transfusion 47:520–528. https://doi.org/10.1111/j.1537-2995.2006.01145.x

    Article  CAS  PubMed  Google Scholar 

  119. Masuyama J, Murakami T, Iwamoto S, Fujita S (2016) Ex vivo expansion of natural killer cells from human peripheral blood mononuclear cells co-stimulated with anti-CD3 and anti-CD52 monoclonal antibodies. Cytotherapy 18:80–90. https://doi.org/10.1016/j.jcyt.2015.09.011

    Article  CAS  PubMed  Google Scholar 

  120. Bachanova V, Burns LJ, McKenna DH, Curtsinger J, Panoskaltsis-Mortari A, Lindgren BR, Cooley S, Weisdorf D, Miller JS (2010) Allogeneic natural killer cells for refractory lymphoma. Cancer Immunol Immunother 59:1739–1744. https://doi.org/10.1007/s00262-010-0896-z

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  121. Lapteva N, Durett AG, Sun J, Rollins LA, Huye LL, Fang J, Dandekar V, Mei Z, Jackson K, Vera J, Ando J, Ngo MC, Coustan-Smith E, Campana D, Szmania S, Garg T, Moreno-Bost A, Vanrhee F, Gee AP, Rooney CM (2012) Large-scale ex vivo expansion and characterization of natural killer cells for clinical applications. Cytotherapy 14:1131–1143. https://doi.org/10.3109/14653249.2012.700767

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  122. Cany J, Dolstra H, Shah N (2015) Umbilical cord blood-derived cellular products for cancer immunotherapy. Cytotherapy 17:739–748. https://doi.org/10.1016/j.jcyt.2015.03.005

    Article  PubMed  Google Scholar 

  123. Eguizabal C, Zenarruzabeitia O, Monge J, Santos S, Vesga MA, Maruri N, Arrieta A, Rinon M, Tamayo-Orbegozo E, Amo L, Larrucea S, Borrego F (2014) Natural killer cells for cancer immunotherapy: pluripotent stem cells-derived NK cells as an immunotherapeutic perspective. Front Immunol 5:439. https://doi.org/10.3389/fimmu.2014.00439

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  124. Nayyar G, Chu Y, Cairo MS (2019) Overcoming resistance to natural killer cell based immunotherapies for solid tumors. Front Oncol 9:51. https://doi.org/10.3389/fonc.2019.00051

    Article  PubMed  PubMed Central  Google Scholar 

  125. Suck G, Odendahl M, Nowakowska P, Seidl C, Wels WS, Klingemann HG, Tonn T (2016) NK-92: an ‘off-the-shelf therapeutic’ for adoptive natural killer cell-based cancer immunotherapy. Cancer Immunol Immunother 65:485–492. https://doi.org/10.1007/s00262-015-1761-x

    Article  CAS  PubMed  Google Scholar 

  126. Koepsell SA, Miller JS, McKenna DH Jr (2013) Natural killer cells: a review of manufacturing and clinical utility. Transfusion 53:404–410. https://doi.org/10.1111/j.1537-2995.2012.03724.x

    Article  CAS  PubMed  Google Scholar 

  127. Dykes JH, Toporski J, Juliusson G, Bekassy AN, Lenhoff S, Lindmark A, Scheding S (2007) Rapid and effective CD3 T-cell depletion with a magnetic cell sorting program to produce peripheral blood progenitor cell products for haploidentical transplantation in children and adults. Transfusion 47:2134–2142. https://doi.org/10.1111/j.1537-2995.2007.01438.x

    Article  PubMed  Google Scholar 

  128. Koehl U, Brehm C, Huenecke S, Zimmermann SY, Kloess S, Bremm M, Ullrich E, Soerensen J, Quaiser A, Erben S, Wunram C, Gardlowski T, Auth E, Tonn T, Seidl C, Meyer-Monard S, Stern M, Passweg J, Klingebiel T, Bader P, Schwabe D, Esser R (2013) Clinical grade purification and expansion of NK cell products for an optimized manufacturing protocol. Front Oncol 3:118. https://doi.org/10.3389/fonc.2013.00118

    Article  PubMed  PubMed Central  Google Scholar 

  129. Ferrara JL, Levine JE, Reddy P, Holler E (2009) Graft-versus-host disease. Lancet 373:1550–1561. https://doi.org/10.1016/S0140-6736(09)60237-3

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  130. Chan YLT, Zuo J, Inman C, Croft W, Begum J, Croudace J, Kinsella F, Maggs L, Nagra S, Nunnick J, Abbotts B, Craddock C, Malladi R, Moss P (2018) NK cells produce high levels of IL-10 early after allogeneic stem cell transplantation and suppress development of acute GVHD. Eur J Immunol 48:316–329. https://doi.org/10.1002/eji.201747134

    Article  CAS  PubMed  Google Scholar 

  131. Oyer JL, Igarashi RY, Kulikowski AR, Colosimo DA, Solh MM, Zakari A, Khaled YA, Altomare DA, Copik AJ (2015) Generation of highly cytotoxic natural killer cells for treatment of acute myelogenous leukemia using a feeder-free, particle-based approach. Biol Blood Marrow Transplant 21:632–639. https://doi.org/10.1016/j.bbmt.2014.12.037

    Article  CAS  PubMed  Google Scholar 

  132. Lapteva N, Szmania SM, van Rhee F, Rooney CM (2014) Clinical grade purification and expansion of natural killer cells. Crit Rev Oncog 19:121–132. https://doi.org/10.1615/CritRevOncog.2014010931

    Article  PubMed  PubMed Central  Google Scholar 

  133. Zwirner NW, Domaica CI (2010) Cytokine regulation of natural killer cell effector functions. BioFactors 36:274–288. https://doi.org/10.1002/biof.107

    Article  CAS  PubMed  Google Scholar 

  134. Freund-Brown J, Chirino L, Kambayashi T (2018) Strategies to enhance NK cell function for the treatment of tumors and infections. Crit Rev Immunol 38:105–130. https://doi.org/10.1615/CritRevImmunol.2018025248

    Article  PubMed  PubMed Central  Google Scholar 

  135. Romee R, Leong JW, Fehniger TA (2014) Utilizing cytokines to function-enable human NK cells for the immunotherapy of cancer. Scientifica (Cairo) 2014:205796. https://doi.org/10.1155/2014/205796

    Article  CAS  PubMed Central  Google Scholar 

  136. Swann JB, Hayakawa Y, Zerafa N, Sheehan KC, Scott B, Schreiber RD, Hertzog P, Smyth MJ (2007) Type I IFN contributes to NK cell homeostasis, activation, and antitumor function. J Immunol 178:7540–7549. https://doi.org/10.4049/jimmunol.178.12.7540

    Article  CAS  PubMed  Google Scholar 

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

    Article  CAS  PubMed  Google Scholar 

  138. Grimm EA, Mazumder A, Zhang HZ, Rosenberg SA (1982) Lymphokine-activated killer cell phenomenon. Lysis of natural killer-resistant fresh solid tumor cells by interleukin 2-activated autologous human peripheral blood lymphocytes. J Exp Med 155:1823–1841. https://doi.org/10.1084/jem.155.6.1823

    Article  CAS  PubMed  Google Scholar 

  139. Miller JS, Soignier Y, Panoskaltsis-Mortari A, McNearney SA, Yun GH, Fautsch SK, McKenna D, Le C, DeFor TE, Burns LJ, Orchard PJ, Blazar BR, Wagner JE, Slungaard A, Weisdorf DJ, Okazaki IJ, McGlave PB (2005) Successful adoptive transfer and in vivo expansion of human haploidentical NK cells in patients with cancer. Blood 105:3051–3057. https://doi.org/10.1182/blood-2004-07-2974

    Article  CAS  PubMed  Google Scholar 

  140. Burns LJ, Weisdorf DJ, DeFor TE, Vesole DH, Repka TL, Blazar BR, Burger SR, Panoskaltsis-Mortari A, Keever-Taylor CA, Zhang MJ, Miller JS (2003) IL-2-based immunotherapy after autologous transplantation for lymphoma and breast cancer induces immune activation and cytokine release: a phase I/II trial. Bone Marrow Transplant 32:177–186. https://doi.org/10.1038/sj.bmt.1704086

    Article  CAS  PubMed  Google Scholar 

  141. Iliopoulou EG, Kountourakis P, Karamouzis MV, Doufexis D, Ardavanis A, Baxevanis CN, Rigatos G, Papamichail M, Perez SA (2010) A phase I trial of adoptive transfer of allogeneic natural killer cells in patients with advanced non-small cell lung cancer. Cancer Immunol Immunother 59:1781–1789. https://doi.org/10.1007/s00262-010-0904-3

    Article  PubMed  Google Scholar 

  142. Mao Y, van Hoef V, Zhang X, Wennerberg E, Lorent J, Witt K, Masvidal L, Liang S, Murray S, Larsson O, Kiessling R, Lundqvist A (2016) IL-15 activates mTOR and primes stress-activated gene expression leading to prolonged antitumor capacity of NK cells. Blood 128:1475–1489. https://doi.org/10.1182/blood-2016-02-698027

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  143. Pillet AH, Bugault F, Theze J, Chakrabarti LA, Rose T (2009) A programmed switch from IL-15- to IL-2-dependent activation in human NK cells. J Immunol 182:6267–6277. https://doi.org/10.4049/jimmunol.0801933

    Article  CAS  PubMed  Google Scholar 

  144. Skak K, Frederiksen KS, Lundsgaard D (2008) Interleukin-21 activates human natural killer cells and modulates their surface receptor expression. Immunology 123:575–583. https://doi.org/10.1111/j.1365-2567.2007.02730.x

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  145. Boyiadzis M, Memon S, Carson J, Allen K, Szczepanski MJ, Vance BA, Dean R, Bishop MR, Gress RE, Hakim FT (2008) Up-regulation of NK cell activating receptors following allogeneic hematopoietic stem cell transplantation under a lymphodepleting reduced intensity regimen is associated with elevated IL-15 levels. Biol Blood Marrow Transplant 14:290–300. https://doi.org/10.1016/j.bbmt.2007.12.490

    Article  CAS  PubMed  Google Scholar 

  146. van Ostaijen-ten Dam MM, Prins HJ, Boerman GH, Vervat C, Pende D, Putter H, Lankester A, van Tol MJ, Zwaginga JJ, Schilham MW (2016) Preparation of cytokine-activated nk cells for use in adoptive cell therapy in cancer patients: protocol optimization and therapeutic potential. J Immunother 39:90–100. https://doi.org/10.1097/CJI.0000000000000110

    Article  CAS  PubMed  Google Scholar 

  147. Kobayashi M, Fitz L, Ryan M, Hewick RM, Clark SC, Chan S, Loudon R, Sherman F, Perussia B, Trinchieri G (1989) Identification and purification of natural killer cell stimulatory factor (NKSF), a cytokine with multiple biologic effects on human lymphocytes. J Exp Med 170:827–845. https://doi.org/10.1084/jem.170.3.827

    Article  CAS  PubMed  Google Scholar 

  148. Ni J, Miller M, Stojanovic A, Garbi N, Cerwenka A (2012) Sustained effector function of IL-12/15/18-preactivated NK cells against established tumors. J Exp Med 209:2351–2365. https://doi.org/10.1084/jem.20120944

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  149. Ni J, Holsken O, Miller M, Hammer Q, Luetke-Eversloh M, Romagnani C, Cerwenka A (2016) Adoptively transferred natural killer cells maintain long-term antitumor activity by epigenetic imprinting and CD4(+) T cell help. Oncoimmunology 5:e1219009. https://doi.org/10.1080/2162402X.2016.1219009

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  150. Ahn YO, Kim S, Kim TM, Song EY, Park MH, Heo DS (2013) Irradiated and activated autologous PBMCs induce expansion of highly cytotoxic human NK cells in vitro. J Immunother 36:373–381. https://doi.org/10.1097/CJI.0b013e3182a3430f

    Article  CAS  PubMed  Google Scholar 

  151. Carlens S, Gilljam M, Chambers BJ, Aschan J, Guven H, Ljunggren HG, Christensson B, Dilber MS (2001) A new method for in vitro expansion of cytotoxic human CD3 CD56+ natural killer cells. Hum Immunol 62:1092–1098. https://doi.org/10.1016/S0198-8859(01)00313-5

    Article  CAS  PubMed  Google Scholar 

  152. Granzin M, Wagner J, Kohl U, Cerwenka A, Huppert V, Ullrich E (2017) Shaping of natural killer cell antitumor activity by ex vivo cultivation. Front Immunol 8:458. https://doi.org/10.3389/fimmu.2017.00458

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  153. Bae DS, Lee JK (2014) Development of NK cell expansion methods using feeder cells from human myelogenous leukemia cell line. Blood Res 49:154–161. https://doi.org/10.5045/br.2014.49.3.154

    Article  PubMed  PubMed Central  Google Scholar 

  154. Alici E, Sutlu T, Bjorkstrand B, Gilljam M, Stellan B, Nahi H, Quezada HC, Gahrton G, Ljunggren HG, Dilber MS (2008) Autologous antitumor activity by NK cells expanded from myeloma patients using GMP-compliant components. Blood 111:3155–3162. https://doi.org/10.1182/blood-2007-09-110312

    Article  CAS  PubMed  Google Scholar 

  155. Barkholt L, Alici E, Conrad R, Sutlu T, Gilljam M, Stellan B, Christensson B, Guven H, Bjorkstrom NK, Soderdahl G, Cederlund K, Kimby E, Aschan J, Ringden O, Ljunggren HG, Dilber MS (2009) Safety analysis of ex vivo-expanded NK and NK-like T cells administered to cancer patients: a phase I clinical study. Immunotherapy 1:753–764. https://doi.org/10.2217/imt.09.47

    Article  CAS  PubMed  Google Scholar 

  156. Sutlu T, Alici E (2011) Ex vivo expansion of natural killer cells: a question of function. Cytotherapy 13:767–768. https://doi.org/10.3109/14653249.2011.563295

    Article  PubMed  Google Scholar 

  157. Miller JS, Oelkers S, Verfaillie C, McGlave P (1992) Role of monocytes in the expansion of human activated natural killer cells. Blood 80:2221–2229

    CAS  PubMed  Google Scholar 

  158. Perussia B, Ramoni C, Anegon I, Cuturi MC, Faust J, Trinchieri G (1987) Preferential proliferation of natural killer cells among peripheral blood mononuclear cells cocultured with B lymphoblastoid cell lines. Nat Immun Cell Growth Regul 6:171–188

    CAS  PubMed  Google Scholar 

  159. Sakamoto N, Ishikawa T, Kokura S, Okayama T, Oka K, Ideno M, Sakai F, Kato A, Tanabe M, Enoki T, Mineno J, Naito Y, Itoh Y, Yoshikawa T (2015) Phase I clinical trial of autologous NK cell therapy using novel expansion method in patients with advanced digestive cancer. J Transl Med 13:277. https://doi.org/10.1186/s12967-015-0632-8

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  160. Imai C, Iwamoto S, Campana D (2005) Genetic modification of primary natural killer cells overcomes inhibitory signals and induces specific killing of leukemic cells. Blood 106:376–383. https://doi.org/10.1182/blood-2004-12-4797

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  161. Fujisaki H, Kakuda H, Shimasaki N, Imai C, Ma J, Lockey T, Eldridge P, Leung WH, Campana D (2009) Expansion of highly cytotoxic human natural killer cells for cancer cell therapy. Cancer Res 69:4010–4017. https://doi.org/10.1158/0008-5472.CAN-08-3712

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  162. Voskens CJ, Watanabe R, Rollins S, Campana D, Hasumi K, Mann DL (2010) Ex-vivo expanded human NK cells express activating receptors that mediate cytotoxicity of allogeneic and autologous cancer cell lines by direct recognition and antibody directed cellular cytotoxicity. J Exp Clin Cancer Res 29:134. https://doi.org/10.1186/1756-9966-29-134

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  163. Caunday O, Bensoussan D, Decot V, Bordigoni P, Stoltz JF (2009) Regulatory aspects of cellular therapy product in Europe: JACIE accreditation in a processing facility. Biomed Mater Eng 19:373–379

    PubMed  Google Scholar 

  164. Sadelain M, Brentjens R, Riviere I (2013) The basic principles of chimeric antigen receptor design. Cancer Discov 3:388–398. https://doi.org/10.1158/2159-8290.CD-12-0548

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  165. Kohl U, Arsenieva S, Holzinger A, Abken H (2018) CAR T cells in trials: recent achievements and challenges that remain in the production of modified T cells for clinical applications. Hum Gene Ther 29:559–568. https://doi.org/10.1089/hum.2017.254

    Article  CAS  PubMed  Google Scholar 

  166. Kalos M, Levine BL, Porter DL, Katz S, Grupp SA, Bagg A, June CH (2011) T cells with chimeric antigen receptors have potent antitumor effects and can establish memory in patients with advanced leukemia. Sci Transl Med 3:95ra73. https://doi.org/10.1126/scitranslmed.3002842

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  167. Hu Y, Tian Z, Zhang C (2019) Natural killer cell-based immunotherapy for cancer: advances and prospects. Engineering 5:106–114. https://doi.org/10.1016/j.eng.2018.11.015

    Article  CAS  Google Scholar 

  168. Bonifant CL, Jackson HJ, Brentjens RJ, Curran KJ (2016) Toxicity and management in CAR T-cell therapy. Mol Ther Oncolytics 3:16011. https://doi.org/10.1038/mto.2016.11

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  169. Shimabukuro-Vornhagen A, Godel P, Subklewe M, Stemmler HJ, Schlosser HA, Schlaak M, Kochanek M, Boll B, von Bergwelt-Baildon MS (2018) Cytokine release syndrome. J Immunother Cancer 6:56. https://doi.org/10.1186/s40425-018-0343-9

    Article  PubMed  PubMed Central  Google Scholar 

  170. Rubnitz JE, Inaba H, Kang G, Gan K, Hartford C, Triplett BM, Dallas M, Shook D, Gruber T, Pui CH, Leung W (2015) Natural killer cell therapy in children with relapsed leukemia. Pediatr Blood Cancer 62:1468–1472. https://doi.org/10.1002/pbc.25555

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  171. Nguyen R, Wu H, Pounds S, Inaba H, Ribeiro RC, Cullins D, Rooney B, Bell T, Lacayo NJ, Heym K, Degar B, Schiff D, Janssen WE, Triplett B, Pui CH, Leung W, Rubnitz JE (2019) A phase II clinical trial of adoptive transfer of haploidentical natural killer cells for consolidation therapy of pediatric acute myeloid leukemia. J Immunother Cancer 7:81. https://doi.org/10.1186/s40425-019-0564-6

    Article  PubMed  PubMed Central  Google Scholar 

  172. Lee DA, Denman CJ, Rondon G, Woodworth G, Chen J, Fisher T, Kaur I, Fernandez-Vina M, Cao K, Ciurea S, Shpall EJ, Champlin RE (2016) Haploidentical natural killer cells infused before allogeneic stem cell transplantation for myeloid malignancies: a phase I trial. Biol Blood Marrow Transplant 22:1290–1298. https://doi.org/10.1016/j.bbmt.2016.04.009

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  173. Bjorklund AT, Carlsten M, Sohlberg E, Liu LL, Clancy T, Karimi M, Cooley S, Miller JS, Klimkowska M, Schaffer M, Watz E, Wikstrom K, Blomberg P, Wahlin BE, Palma M, Hansson L, Ljungman P, Hellstrom-Lindberg E, Ljunggren HG, Malmberg KJ (2018) Complete remission with reduction of high-risk clones following haploidentical NK-cell therapy against MDS and AML. Clin Cancer Res 24:1834–1844

    CAS  PubMed  Google Scholar 

  174. Munoz BM, Vela CM, Fuster Soler JL, Astigarraga I, Pascual MA, Vagace Valero JM, Tong HY, Valentin QJ, Fernandez CL, Escudero LA, Sisinni L, Blanquer M, Mirones AI, Gonzalez MB, Borobia AM, Perez-Martinez A (2020) Study protocol for a phase II, multicentre, prospective, non-randomised clinical trial to assess the safety and efficacy of infusing allogeneic activated and expanded natural killer cells as consolidation therapy for paediatric acute myeloblastic leukaemia. BMJ Open 10:e029642

    Google Scholar 

  175. Cooley S, He F, Bachanova V, Vercellotti GM, DeFor TE, Curtsinger JM, Robertson P, Grzywacz B, Conlon KC, Waldmann TA, McKenna DH, Blazar BR, Weisdorf DJ, Miller JS (2019) First-in-human trial of rhIL-15 and haploidentical natural killer cell therapy for advanced acute myeloid leukemia. Blood Adv 3:1970–1980. https://doi.org/10.1182/bloodadvances.2018028332

    Article  PubMed  PubMed Central  Google Scholar 

  176. Szmania S, Lapteva N, Garg T, Greenway A, Lingo J, Nair B, Stone K, Woods E, Khan J, Stivers J, Panozzo S, Campana D, Bellamy WT, Robbins M, Epstein J, Yaccoby S, Waheed S, Gee A, Cottler-Fox M, Rooney C, Barlogie B, van Rhee F (2015) Ex vivo-expanded natural killer cells demonstrate robust proliferation in vivo in high-risk relapsed multiple myeloma patients. J Immunother 38:24–36. https://doi.org/10.1097/CJI.0000000000000059

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  177. Bachanova V, Sarhan D, DeFor TE, Cooley S, Panoskaltsis-Mortari A, Blazar BR, Curtsinger JM, Burns L, Weisdorf DJ, Miller JS (2018) Haploidentical natural killer cells induce remissions in non-Hodgkin lymphoma patients with low levels of immune-suppressor cells. Cancer Immunol Immunother 67:483–494. https://doi.org/10.1007/s00262-017-2100-1

    Article  CAS  PubMed  Google Scholar 

  178. Adotevi O, Godet Y, Galaine J, Lakkis Z, Idirene I, Certoux JM, Jary M, Loyon R, Laheurte C, Kim S, Dormoy A, Pouthier F, Barisien C, Fein F, Tiberghien P, Pivot X, Valmary-Degano S, Ferrand C, Morel P, Delabrousse E, Borg C (2018) In situ delivery of allogeneic natural killer cell (NK) combined with Cetuximab in liver metastases of gastrointestinal carcinoma: a phase I clinical trial. Oncoimmunology 7:e1424673. https://doi.org/10.1080/2162402X.2018.1424673

    Article  CAS  PubMed  PubMed Central  Google Scholar 

Download references

Funding

This work was supported by: Instituto de Salud Carlos III, Spain (PI13/02691 to Rafael Solana and PI16/01615 to Rafael Solana and Corona Alonso). Agencia Estatal de Investigación, Ministry of Economy and Competitiveness of Spain (SAF2013-46161-R and SAF2017-87538-R to Raquel Tarazona). Consejería de Economía e Infraestructuras, Junta de Extremadura (IB16164 and grants to INPATT (CTS040) research group GR18085 to Raquel Tarazona). Grants were cofinanced by European Regional Development Funds (FEDER) “Una manera de hacer Europa”. Grant TE-0039-18 (to Beatriz Guerrero) from Consejería de Educación y Empleo cofinanced by European Social Fund, Youth Employment Initiative, “El FSE invierte en tu futuro”. Plan Propio Universidad de Córdoba (to Alejandra Pera).

Author information

Authors and Affiliations

Authors

Contributions

RT, CA and RS designed the first draft of the manuscript. NL, BG, FH and IV contributed to the writing of different sections, figures and tables. AP, BS, NP and ED discussed the manuscript sections and contributed with updated references. All authors revised and agreed to the final version of the paper.

Corresponding authors

Correspondence to Raquel Tarazona, Corona Alonso or Rafael Solana.

Ethics declarations

Conflict of interest

The authors declare that they have no conflict of interest.

Additional information

Publisher's Note

Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Tarazona, R., Lopez-Sejas, N., Guerrero, B. et al. Current progress in NK cell biology and NK cell-based cancer immunotherapy. Cancer Immunol Immunother 69, 879–899 (2020). https://doi.org/10.1007/s00262-020-02532-9

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1007/s00262-020-02532-9

Keywords

Navigation