614
Views
16
CrossRef citations to date
0
Altmetric
Review

The rise of human stem cell-derived natural killer cells for cancer immunotherapy

, , ORCID Icon &
Pages 141-148 | Received 18 Oct 2018, Accepted 11 Dec 2018, Published online: 24 Dec 2018

References

  • Morvan MG, Lanier LL. NK cells and cancer: you can teach innate cells new tricks. Nat Rev Cancer. 2016;16:7–19.
  • Vivier E, Raulet DH, Moretta A, et al. Innate or adaptive immunity? The example of natural killer cells. Science. 2011;331:44–49.
  • Jacobs R, Hintzen G, Kemper A, et al. CD56(bright) cells differ in their KIR repertoire and cytotoxic features from CD56(dim) NK cells. Eur J Immunol. 2001;31:3121–3127.
  • Malmberg KJ, Carlsten M, Bjorklund A, et al. Natural killer cell-mediated immunosurveillance of human cancer. Semin Immunol. 2017;31:20–29.
  • Long EO, Kim HS, Liu D, et al. Controlling natural killer cell responses: integration of signals for activation and inhibition. Annu Rev Immunol. 2013;31:227–258.
  • Ljunggren HG, Karre K. In search of the ‘missing self’: MHC molecules and NK cell recognition. Immunol Today. 1990;11:237–244.
  • Raulet DH, Vance RE. Self-tolerance of natural killer cells. Nat Rev Immunol. 2006;6:520–531.
  • Martinez-Lostao L, Anel A, Pardo J. How do cytotoxic lymphocytes kill cancer cells? Clin Cancer Res. 2015;21:5047–5056.
  • Zamai L, Ahmad M, Bennett IM, et al. Natural killer (NK) cell-mediated cytotoxicity: differential use of TRAIL and Fas ligand by immature and mature primary human NK cells. J Exp Med. 1998;188:2375–2380.
  • Romee R, Foley B, Lenvik T, et al. NK cell CD16 surface expression and function is regulated by a disintegrin and metalloprotease-17 (ADAM17). Blood. 2013;121:3599–3608.
  • Jing Y, Ni Z, Wu J, et al. Identification of an ADAM17 cleavage region in human CD16 (FcgammaRIII) and the engineering of a non-cleavable version of the receptor in NK cells. PLoS One. 2015;10:e0121788.
  • Muller L, Aigner P, Stoiber D. Type I interferons and natural killer cell regulation in cancer. Front Immunol. 2017;8:304.
  • Hartmann J, Schussler-Lenz M, Bondanza A, et al. Clinical development of CAR T cells-challenges and opportunities in translating innovative treatment concepts. EMBO Mol Med. 2017;9:1183–1197.
  • Lim WA, June CH. The principles of engineering immune cells to treat cancer. Cell. 2017;168:724–740.
  • June CH, O’Connor RS, Kawalekar OU, et al. CAR T cell immunotherapy for human cancer. Science. 2018;359:1361–1365.
  • Ruella M, Kenderian SS. Next-generation chimeric antigen receptor T-cell therapy: going off the shelf. BioDrugs. 2017;31:473–481.
  • Cooper ML, Choi J, Staser K, et al. An “off-the-shelf” fratricide-resistant CAR-T for the treatment of T cell hematologic malignancies. Leukemia. 2018;32:1970–1983.
  • Miller JS, Soignier Y, Panoskaltsis-Mortari A, et al. Successful adoptive transfer and in vivo expansion of human haploidentical NK cells in patients with cancer. Blood. 2005;105:3051–3057.
  • Geller MA, Cooley S, Judson PL, et al. A phase II study of allogeneic natural killer cell therapy to treat patients with recurrent ovarian and breast cancer. Cytotherapy. 2011;13:98–107.
  • Dolstra H, Roeven MWH, Spanholtz J, et al. Successful transfer of umbilical cord blood CD34(+) hematopoietic stem and progenitor-derived NK cells in older acute myeloid leukemia patients. Clin Cancer Res. 2017;23:4107–4118.
  • Romee R, Rosario M, Berrien-Elliott MM, et al. Cytokine-induced memory-like natural killer cells exhibit enhanced responses against myeloid leukemia. Sci Transl Med. 2016;8:357ra123.
  • Della Chiesa M, Falco M, Podesta M, et al. Phenotypic and functional heterogeneity of human NK cells developing after umbilical cord blood transplantation: a role for human cytomegalovirus? Blood. 2012;119:399–410.
  • Ciurea SO, Schafer JR, Bassett R, et al. Phase 1 clinical trial using mbIL21 ex vivo-expanded donor-derived NK cells after haploidentical transplantation. Blood. 2017;130:1857–1868.
  • Zhu H, Lai YS, Li Y, et al. Concise review: human Pluripotent stem cells to produce cell-based cancer immunotherapy. Stem Cells. 2018;36:134–145.
  • Sarvaria A, Jawdat D, Madrigal JA, et al. Umbilical cord blood natural killer cells, their characteristics, and potential clinical applications. Front Immunol. 2017;8:329.
  • Woll PS, Martin CH, Miller JS, et al. Human embryonic stem cell-derived NK cells acquire functional receptors and cytolytic activity. J Immunol. 2005;175:5095–5103.
  • Woll PS, Grzywacz B, Tian X, et al. Human embryonic stem cells differentiate into a homogeneous population of natural killer cells with potent in vivo antitumor activity. Blood. 2009;113:6094–6101.
  • Knorr DA, Ni Z, Hermanson D, et al. Clinical-scale derivation of natural killer cells from human pluripotent stem cells for cancer therapy. Stem Cells Transl Med. 2013;2:274–283.
  • Bock AM, Knorr D, Kaufman DS. Development, expansion, and in vivo monitoring of human NK cells from human embryonic stem cells (hESCs) and and induced pluripotent stem cells (iPSCs). J Vis Exp. 2013;74:e50337.
  • Sakamoto N, Ishikawa T, Kokura S, et al. Phase I clinical trial of autologous NK cell therapy using novel expansion method in patients with advanced digestive cancer. J Transl Med. 2015;13:277.
  • Parkhurst MR, Riley JP, Dudley ME, et al. 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. 2011;17:6287–6297.
  • Veluchamy JP, Kok N, van der Vliet HJ, et al. The rise of allogeneic natural killer cells as a platform for cancer immunotherapy: recent innovations and future developments. Front Immunol. 2017;8.
  • Dahlberg CI, Sarhan D, Chrobok M, et al. Natural killer cell-based therapies targeting cancer: possible strategies to gain and sustain anti-tumor activity. Front Immunol. 2015;6:605.
  • Barkholt L, Alici E, Conrad R, et al. Safety analysis of ex vivo-expanded NK and NK-like T cells administered to cancer patients: a phase I clinical study. Immunotherapy-Uk. 2009;1:753–764.
  • Iliopoulou EG, Kountourakis P, Karamouzis MV, et al. A phase I trial of adoptive transfer of allogeneic natural killer cells in patients with advanced non-small cell lung cancer. Cancer Immunol Immunother. 2010;59:1781–1789.
  • Veluchamy JP, Kok N, van der Vliet HJ, et al. The rise of allogeneic natural killer cells as a platform for cancer immunotherapy: recent innovations and future developments. Front Immunol. 2017;8:631.
  • Handgretinger R, Lang P, Andre MC. Exploitation of natural killer cells for the treatment of acute leukemia. Blood. 2016;127:3341–3349.
  • Liu E, Tong Y, Dotti G, et al. Cord blood NK cells engineered to express IL-15 and a CD19-targeted CAR show long-term persistence and potent antitumor activity. Leukemia. 2018;32:520–531.
  • Williams BA, Law AD, Routy B, et al. A phase I trial of NK-92 cells for refractory hematological malignancies relapsing after autologous hematopoietic cell transplantation shows safety and evidence of efficacy. Oncotarget. 2017;8:89256–89268.
  • Passweg JR, Tichelli A, Meyer-Monard S, et al. Purified donor NK-lymphocyte infusion to consolidate engraftment after haploidentical stem cell transplantation. Leukemia. 2004;18:1835–1838.
  • Carlsten M, Childs RW. Genetic manipulation of NK cells for cancer immunotherapy: techniques and clinical implications. Front Immunol. 2015;6:266.
  • Ruella M, Xu J, Barrett DM, et al. Induction of resistance to chimeric antigen receptor T cell therapy by transduction of a single leukemic B cell. Nat Med. 2018;24:1499–1503.
  • Tonn T, Schwabe D, Klingemann HG, et al. Treatment of patients with advanced cancer with the natural killer cell line NK-92. Cytotherapy. 2013;15:1563–1570.
  • Klingemann H, Boissel L, Toneguzzo F. Natural killer cells for immunotherapy – advantages of the NK-92 cell line over blood NK cells. Front Immunol. 2016;7:91.
  • Zhang C, Oberoi P, Oelsner S, et al. Chimeric antigen receptor-engineered NK-92 cells: an off-the-shelf cellular therapeutic for targeted elimination of cancer cells and induction of protective antitumor immunity. Front Immunol. 2017;8:533.
  • Clemenceau B, Vivien R, Pellat C, et al. The human natural killer cytotoxic cell line NK-92, once armed with a murine CD16 receptor, represents a convenient cellular tool for the screening of mouse mAbs according to their ADCC potential. mAbs. 2013;5:587–594.
  • Li Y, Hermanson DL, Moriarity BS, et al. Human iPSC-derived natural killer cells engineered with chimeric antigen receptors enhance anti-tumor activity. Cell Stem Cell. 2018;23:181–92 e5.
  • Wilber A, Linehan JL, Tian X, et al. Efficient and stable transgene expression in human embryonic stem cells using transposon-mediated gene transfer. Stem Cells. 2007;25:2919–2927.
  • Johnson JK, Miller JS. Current strategies exploiting NK-cell therapy to treat haematologic malignancies. Int J Immunogenet. 2018; doi: 10.1111/iji.12387.
  • Hermanson DL, Ni Z, Kaufman DS. Human pluripotent stem cells as a renewable source of natural killer cells. In: Cheng T, editor. Hematopoietic differentiation of human pluripotent stem cells. Dordrecht: Springer Netherlands; 2015. p. 69–79.
  • Knorr DA, Bock A, Brentjens RJ, et al. Engineered human embryonic stem cell-derived lymphocytes to study in vivo trafficking and immunotherapy. Stem Cells Dev. 2013;22:1861–1869.
  • Hermanson DL. Human pluripotent stem cells as a renewable source of natural killer cells. NK book chapter; 2015.
  • Hermanson DL, Bendzick L, Pribyl L, et al. Induced pluripotent stem cell-derived natural killer cells for treatment of ovarian cancer. Stem Cells. 2016;34:93–101.
  • Denman CJ, Senyukov VV, Somanchi SS, et al. Membrane-bound IL-21 promotes sustained ex vivo proliferation of human natural killer cells. PLoS One. 2012;7:e30264.
  • Ni Z, Knorr DA, Bendzick L, et al. Expression of chimeric receptor CD4zeta by natural killer cells derived from human pluripotent stem cells improves in vitro activity but does not enhance suppression of HIV infection in vivo. Stem Cells. 2014;32:1021–1031.
  • Burga RA, Nguyen T, Zulovich J, et al. Improving efficacy of cancer immunotherapy by genetic modification of natural killer cells. Cytotherapy. 2016;18:1410–1421.
  • Murray S, Lundqvist A. Targeting the tumor microenvironment to improve natural killer cell-based immunotherapies: on being in the right place at the right time, with resilience. Hum Vaccine Immunother. 2016;12:607–611.
  • Vitale M, Cantoni C, Pietra G, et al. Effect of tumor cells and tumor microenvironment on NK-cell function. Eur J Immunol. 2014;44:1582–1592.
  • Ghiringhelli F, Menard C, Terme M, et al. CD4(+) CD25(+) regulatory T cells inhibit natural killer cell functions in a transforming growth factor-beta-dependent manner. J Exp Med. 2005;202:1075–1085.
  • Beavis PA, Divisekera U, Paget C, et al. Blockade of A2A receptors potently suppresses the metastasis of CD73+ tumors. Proc Natl Acad Sci USA. 2013;110:14711–14716.
  • Kai S, Goto S, Tahara K, et al. Inhibition of indoleamine 2,3-dioxygenase suppresses NK cell activity and accelerates tumor growth. J Exp Ther Oncol. 2003;3:336–345.
  • Fauriat C, Just-Landi S, Mallet F, et al. Deficient expression of NCR in NK cells from acute myeloid leukemia: evolution during leukemia treatment and impact of leukemia cells in NCRdull phenotype induction. Blood. 2007;109:323–330.
  • Carlsten M, Norell H, Bryceson YT, et al. Primary human tumor cells expressing CD155 impair tumor targeting by down-regulating DNAM-1 on NK cells. J Iimmunol. 2009;183:4921–4930.
  • Morgado S, Sanchez-Corre B, Casado JG, et al. NK cell recognition and killing of melanoma cells is controlled by multiple activating receptor-ligand interactions. J Innate Immun. 2011;3:365–373.
  • Sanchez-Correa B, Gayoso I, Bergua JM, et al. Decreased expression of DNAM-1 on NK cells from acute myeloid leukemia patients. Immunol Cell Biol. 2012;90:109–115.
  • Huntington ND, Legrand N, Alves NL, et al. IL-15 trans-presentation promotes human NK cell development and differentiation in vivo. J Exp Med. 2009;206:25–34.
  • Lehmann C, Zeis M, Uharek L. Activation of natural killer cells with interleukin 2 (IL-2) and IL-12 increases perforin binding and subsequent lysis of tumour cells. Br J Haematol. 2001;114:660–665.
  • Schwartz RN, Stover L, Dutcher J. Managing toxicities of high-dose interleukin-2. Oncology-Ny. 2002;16:11–20.
  • Berger C, Berger M, Hackman RC, et al. Safety and immunologic effects of IL-15 administration in nonhuman primates. Blood. 2009;114:2417–2426.
  • Sanchez-Correa B, Bergua JM, Pera A, et al. 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. 2017;8:931.
  • Romee R, Cooley S, Berrien-Elliott MM, et al. First-in-human phase 1 clinical study of the IL-15 superagonist complex ALT-803 to treat relapse after transplantation. Blood. 2018;131:2515–2527.
  • Mathios D, Park CK, Marcus WD, et al. Therapeutic administration of IL-15 superagonist complex ALT-803 leads to long-term survival and durable antitumor immune response in a murine glioblastoma model. Int J Cancer. 2016;138:187–194.
  • Tamzalit F, Barbieux I, Plet A, et al. IL-15.IL-15R alpha complex shedding following trans-presentation is essential for the survival of IL-15 responding NK and T cells. Proc Natl Acad Sci USA. 2014;111:8565–8570.
  • Mishra A, Sullivan L, Caligiuri MA. Molecular pathways: interleukin-15 signaling in health and in cancer. Clin Cancer Res. 2014;20:2044–2050.
  • Imamura M, Shook D, Kamiya T, et al. Autonomous growth and increased cytotoxicity of natural killer cells expressing membrane-bound interleukin-15. Blood. 2014;124:1081–1088.
  • Hurton LV, Singh H, Najjar AM, et al. Tethered IL-15 augments antitumor activity and promotes a stem-cell memory subset in tumor-specific T cells. Proc Natl Acad Sci USA. 2016;113:E7788–E97.
  • Lee TT, Li YE, Bjordahl R, et al. Abstract 3574: cytokine-autonomous, CAR-directed, off-the-shelf natural killer cells derived from a clonal engineered master pluripotent cell line. Cancer Res. 2018;78:3574.
  • Keating N, Nicholson SE. SOCS-mediated immunomodulation of natural killer cells. Cytokine. 2018; pii:S1043-4666(18)30119-4.
  • Delconte RB, Kolesnik TB, Dagley LF, et al. CIS is a potent checkpoint in NK cell-mediated tumor immunity. Cytokine. 2016;87:153–154.
  • Zhu H, Kaufman DS. CISH deletion combined with NOTCH activation leads to production of hyper-active human natural killer cells derived from human pluripotent stem cells. Exp Hematol. 2018;64:S76.
  • Nimmerjahn F, Ravetch JV. Fc gamma receptors: old friends and new family members. Immunity. 2006;24:19–28.
  • Yeap WH, Wong KL, Shimasaki N, et al. CD16 is indispensable for antibody-dependent cellular cytotoxicity by human monocytes. Sci Rep. 2016;6:34310.
  • Bibeau F, Lopez-Crapez E, Di Fiore F, et al. Impact of FcγRIIa-FcγRIIIa polymorphisms and KRAS mutations on the clinical outcome of patients with metastatic colorectal cancer treated with cetuximab plus irinotecan. J Clin Oncol. 2009;27:1122–1129.
  • Cartron G, Dacheux L, Salles G, et al. Therapeutic activity of humanized anti-CD20 monoclonal antibody and polymorphism in IgG Fc receptor FcγRIIIa gene. Blood. 2002;99:754–758.
  • Musolino A, Naldi N, Bortesi B, et al. Immunoglobulin G fragment C receptor polymorphisms and clinical efficacy of trastuzumab-based therapy in patients with HER-2/neu-positive metastatic breast cancer. J Clin Oncol. 2008;26:1789–1796.
  • Zhou Q, Gil-Krzewska A, Peruzzi G, et al. Matrix metalloproteinases inhibition promotes the polyfunctionality of human natural killer cells in therapeutic antibody-based anti-tumour immunotherapy. Clin Exp Immunol. 2013;173:131–139.
  • Zhu H, Bjordahl R, Gaidarova S, et al. Genetically engineered pluripotent cell-derived natural killer cell therapy provides enhanced antibody dependent cellular cytotoxicity against hematologic malignancies and solid tumors in combination with monoclonal antibody therapy. Blood. 2017;130:4452.
  • Liu DF, Tian S, Zhang K, et al. Chimeric antigen receptor (CAR)-modified natural killer cell-based immunotherapy and immunological synapse formation in cancer and HIV. Protein Cell. 2017;8:861–877.
  • Romanski A, Uherek C, Bug G, et al. CD19-CAR engineered NK-92 cells are sufficient to overcome NK cell resistance in B-cell malignancies. J Cell Mol Med. 2016;20:1287–1294.
  • Tang X, Yang L, Li Z, et al. First-in-man clinical trial of CAR NK-92 cells: safety test of CD33-CAR NK-92 cells in patients with relapsed and refractory acute myeloid leukemia. Am J Cancer Res. 2018;8:1083–1089.
  • Bollino D, Webb TJ. Chimeric antigen receptor-engineered natural killer and natural killer T cells for cancer immunotherapy. Transl Res. 2017;187:32–43.
  • Hermanson DL, Kaufman DS. Utilizing chimeric antigen receptors to direct natural killer cell activity. Front Immunol. 2015;6:195.
  • Koehl U, Kalberer C, Spanholtz J, et al. Advances in clinical NK cell studies: donor selection, manufacturing and quality control. Oncoimmunology. 2016;5.

Reprints and Corporate Permissions

Please note: Selecting permissions does not provide access to the full text of the article, please see our help page How do I view content?

To request a reprint or corporate permissions for this article, please click on the relevant link below:

Academic Permissions

Please note: Selecting permissions does not provide access to the full text of the article, please see our help page How do I view content?

Obtain permissions instantly via Rightslink by clicking on the button below:

If you are unable to obtain permissions via Rightslink, please complete and submit this Permissions form. For more information, please visit our Permissions help page.