The War Is on: The Immune System against Glioblastoma—How Can NK Cells Drive This Battle?
Abstract
:1. Introduction
2. Natural Killer Cells
3. NK Cells in Immunosurveillance against Cancer
3.1. Tumor Cell Recognition
3.2. Antitumor Functions
4. NK Cells in GBM
5. NK-Cell-Based Immunotherapy
5.1. Adoptive Cell Therapy
5.2. CAR-NK Cells
5.3. Cytokine Therapy
5.4. Monoclonal Antibodies, Killer Cell Engagers, Immunocytokines, and Other Antibody-Based Strategies to Boost NK Activation
5.5. Tumor Sensitization
6. Conclusions
Author Contributions
Funding
Institutional Review Board Statement
Informed Consent Statement
Conflicts of Interest
References
- Sung, H.; Ferlay, J.; Siegel, R.L.; Laversanne, M.; Soerjomataram, I.; Jemal, A.; Bray, F. Global Cancer Statistics 2020: GLOBOCAN Estimates of Incidence and Mortality Worldwide for 36 Cancers in 185 Countries. CA Cancer J. Clin. 2021, 71, 209–249. [Google Scholar] [CrossRef]
- Wirsching, H.G.; Galanis, E.; Weller, M. Glioblastoma. In Handbook of Clinical Neurology; Berger, M.S., Weller, M., Eds.; Elsevier: Amsterdam, The Netherlands, 2016; Volume 134, pp. 381–397. ISBN 9780128029978. [Google Scholar]
- Stupp, R.; Mason, W.P.; van den Bent, M.J.; Weller, M.; Fisher, B.; Taphoorn, M.J.B.; Belanger, K.; Brandes, A.A.; Marosi, C.; Bogdahn, U.; et al. Radiotherapy plus Concomitant and Adjuvant Temozolomide for Glioblastoma. N. Engl. J. Med. 2005, 352, 987–996. [Google Scholar] [CrossRef] [PubMed]
- Tran, B.; Rosenthal, M.A. Survival comparison between glioblastoma multiforme and other incurable cancers. J. Clin. Neurosci. 2010, 17, 417–421. [Google Scholar] [CrossRef]
- Ostrom, Q.T.; Gittleman, H.; Liao, P.; Rouse, C.; Chen, Y.; Dowling, J.; Wolinsky, Y.; Kruchko, C.; Barnholtz-Sloan, J. CBTRUS statistical report: Primary brain and central nervous system tumors diagnosed in the United States in 2007–2011. Neuro-Oncol. 2014, 16, iv1–iv63. [Google Scholar] [CrossRef] [PubMed]
- Perlmann, P.; Holm, G. Cytotoxic effects of lymphoid cells in vitro. Adv. Immunol. 1969, 11, 117–193. [Google Scholar] [CrossRef]
- Rosenau, W.; Moon, H.D. Lysis of homologous cells by sensitized lymphocytes in tissue culture. J. Natl. Cancer Inst. 1961, 27, 471–483. [Google Scholar] [PubMed]
- Rosenberg, E.; Herberman, R.; Levine, P.; Halterman, R.; McCoy, J.; Wunderlich, J. Lymphocyte cytotoxicity reactions to leukemia-associated antigens in identical twins. Int. J. Cancer 1972, 9, 648–658. [Google Scholar] [CrossRef]
- Ozer, H.; Strelkauskas, A.J.; Callery, R.T.; Schlossman, S.F. The functional dissection of human peripheral null cells with respect to antibody-dependent cellular cytotoxicity and natural killing. Eur. J. Immunol. 1979, 9, 112–118. [Google Scholar] [CrossRef]
- Trinchieri, G. Biology of natural killer cells. Adv. Immunol. 1989, 47, 187–376. [Google Scholar] [CrossRef]
- Perussia, B.; Acuto, O.; Terhorst, C.; Faust, J.; Lazarus, R.; Fanning, V.; Trinchieri, G. Human natural killer cells analyzed by B73.1, a monoclonal antibody blocking Fc receptor functions. II. Studies of B73.1 antibody-antigen interaction on the lymphocyte membrane. J. Immunol. 1983, 130, 2142–2148. [Google Scholar]
- Grossi, C.E.; Cadoni, A.; Zicca, A.; Leprini, A.; Ferrarini, M. Large Granular Lymphocytes in Human Peripheral Blood: Ultrastructural and Cytochemical Characterization of the Granules. Blood 1982, 59, 277–283. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Kiessling, R.; Klein, E.; Wigzell, H. “Natural” killer cells in the mouse. I. Cytotoxic cells with specificity for mouse Moloney leukemia cells. Specificity and distribution according to genotype. Eur. J. Immunol. 1975, 5, 112–117. [Google Scholar] [CrossRef]
- Ljunggren, H.G.; Kärre, K. In search of the “missing self”: MHC molecules and NK cell recognition. Immunol. Today 1990, 11, 237–244. [Google Scholar] [CrossRef]
- Valiante, N.; Lienert, K.; Shilling, H.; Smits, B.; Parham, P. Killer cell receptors: Keeping pace with MHC class I evolution. Immunol. Rev. 1997, 155, 155–164. [Google Scholar] [CrossRef]
- Chiossone, L.; Dumas, P.Y.; Vienne, M.; Vivier, E. Natural killer cells and other innate lymphoid cells in cancer. Nat. Rev. Immunol. 2018, 18, 671–688. [Google Scholar] [CrossRef]
- Xie, G.; Dong, H.; Liang, Y.; Ham, J.D.; Rizwan, R.; Chen, J. CAR-NK cells: A promising cellular immunotherapy for cancer. EBioMedicine 2020, 59, 102975. [Google Scholar] [CrossRef] [PubMed]
- Carrega, P.; Ferlazzo, G. Natural killer cell distribution and trafficking in human tissues. Front. Immunol. 2012, 3, 347. [Google Scholar] [CrossRef] [Green Version]
- Cerwenka, A.; Lanier, L.L. Natural killer cell memory in infection, inflammation and cancer. Nat. Rev. Immunol. 2016, 16, 112–123. [Google Scholar] [CrossRef]
- Walzer, T.; Bléry, M.; Chaix, J.; Fuseri, N.; Chasson, L.; Robbins, S.H.; Jaeger, S.; André, P.; Gauthier, L.; Daniel, L.; et al. Identification, activation, and selective in vivo ablation of mouse NK cells via NKp46. Proc. Natl. Acad. Sci. USA 2007, 104, 3384–3389. [Google Scholar] [CrossRef] [Green Version]
- Carlyle, J.R.; Mesci, A.; Ljutic, B.; Belanger, S.; Tai, L.-H.; Rousselle, E.; Troke, A.D.; Proteau, M.-F.; Makrigiannis, A.P. Molecular and Genetic Basis for Strain-Dependent NK1.1 Alloreactivity of Mouse NK Cells. J. Immunol. 2006, 176, LP-7511–LP-7524. [Google Scholar] [CrossRef] [Green Version]
- Lanier, L.L.; Testi, R.; Bindl, J.; Phillips, J.H. Identity of Leu-19 (CD56) leukocyte differentiation antigen and neural cell adhesion molecule. J. Exp. Med. 1989, 169, 2233–2238. [Google Scholar] [CrossRef] [Green Version]
- Ritz, J.; Schmidt, R.E.; Michon, J.; Hercend, T.; Schlossman, S.F. Characterization of functional surface structures on human natural killer cells. Adv. Immunol. 1988, 42, 181–211. [Google Scholar] [CrossRef] [PubMed]
- Cichocki, F.; Grzywacz, B.; Miller, J.S. Human NK cell development: One road or many? Front. Immunol. 2019, 10, 2078. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Zhang, Y.; Payne, K.J.; Zhu, Y.; Price, M.A.; Parrish, Y.K.; Zielinska, E.; Barsky, L.W.; Crooks, G.M. SCL Expression at Critical Points in Human Hematopoietic Lineage Commitment. Stem Cells 2005, 23, 852–860. [Google Scholar] [CrossRef] [PubMed]
- Renoux, V.; Zriwil, A.; Peitzsch, C.; Michaëlsson, J.; Friberg, D.; Soneji, S.; Sitnicka, E. Identification of a Human Natural Killer Cell Lineage-Restricted Progenitor in Fetal and Adult Tissues. Immunity 2015, 43, 394–407. [Google Scholar] [CrossRef] [Green Version]
- Freud, A.; Yokohama, A.; Becknell, B.; Lee, M.; Mao, H.; Ferketich, A.; Caligiuri, M. Evidence for discrete stages of human natural killer cell differentiation in vivo. J. Exp. Med. 2006, 203, 1033–1043. [Google Scholar] [CrossRef] [Green Version]
- Scoville, S.; Freud, A.; Caligiuri, M. Modeling Human Natural Killer Cell Development in the Era of Innate Lymphoid Cells. Front. Immunol. 2017, 8, 360. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Lyman, S.D.; Jacobsen, S.E. c-kit ligand and Flt3 ligand: Stem/progenitor cell factors with overlapping yet distinct activities. Blood 1998, 91, 1101–1134. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Robin, C.; Ottersbach, K.; Durand, C.; Peeters, M.; Vanes, L.; Tybulewicz, V.; Dzierzak, E. An unexpected role for IL-3 in the embryonic development of hematopoietic stem cells. Dev. Cell 2006, 11, 171–180. [Google Scholar] [CrossRef] [Green Version]
- Yu, H.; Fehniger, T.A.; Fuchshuber, P.; Thiel, K.S.; Vivier, E.; Carson, W.E.; Caligiuri, M.A. Flt3 ligand promotes the generation of a distinct CD34(+) human natural killer cell progenitor that responds to interleukin-15. Blood 1998, 92, 3647–3657. [Google Scholar] [CrossRef] [PubMed]
- Giri, J.G.; Ahdieh, M.; Eisenman, J.; Shanebeck, K.; Grabstein, K.; Kumaki, S.; Namen, A.; Park, L.S.; Cosman, D.; Anderson, D. Utilization of the beta and gamma chains of the IL-2 receptor by the novel cytokine IL-15. EMBO J. 1994, 13, 2822–2830. [Google Scholar] [CrossRef]
- Kennedy, M.; Glaccum, M.; Brown, S.; Butz, E.; Viney, J.; Embers, M.; Matsuki, N.; Charrier, K.; Sedger, L.; Willis, C.; et al. Reversible defects in natural killer and memory CD8 T cell lineages in interleukin 15-deficient mice. J. Exp. Med. 2000, 191, 771–780. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Boos, M.; Ramirez, K.; Kee, B. Extrinsic and intrinsic regulation of early natural killer cell development. Immunol. Res. 2008, 40, 193–207. [Google Scholar] [CrossRef] [PubMed]
- Vosshenrich, C.; Ranson, T.; Samson, S.; Corcuff, E.; Colucci, F.; Rosmaraki, E.; Di Santo, J. Roles for common cytokine receptor gamma-chain-dependent cytokines in the generation, differentiation, and maturation of NK cell precursors and peripheral NK cells in vivo. J. Immunol. 2005, 174, 1213–1221. [Google Scholar] [CrossRef] [PubMed]
- Lodolce, J.; Boone, D.; Chai, S.; Swain, R.; Dassopoulos, T.; Trettin, S.; Ma, A. IL-15 receptor maintains lymphoid homeostasis by supporting lymphocyte homing and proliferation. Immunity 1998, 9, 669–676. [Google Scholar] [CrossRef] [Green Version]
- Rosmaraki, E.E.; Douagi, I.; Roth, C.; Colucci, F.; Cumano, A.; Di Santo, J.P. Identification of committed NK cell progenitors in adult murine bone marrow. Eur. J. Immunol. 2001, 31, 1900–1909. [Google Scholar] [CrossRef]
- Nitta, T.; Yagita, H.; Sato, K.; Okumura, K. Involvement of CD56 (NKH-1/Leu-19 antigen) as an adhesion molecule in natural killer-target cell interaction. J. Exp. Med. 1989, 170, 1757–1761. [Google Scholar] [CrossRef] [PubMed]
- Kim, S.; Iizuka, K.; Kang, H.; Dokun, A.; French, A.; Greco, S.; Yokoyama, W. In vivo developmental stages in murine natural killer cell maturation. Nat. Immunol. 2002, 3, 523–528. [Google Scholar] [CrossRef]
- Bryceson, Y.T.; Chiang, S.C.C.; Darmanin, S.; Fauriat, C.; Schlums, H.; Theorell, J.; Wood, S.M. Molecular mechanisms of natural killer cell activation. J. Innate Immun. 2011, 3, 216–226. [Google Scholar] [CrossRef]
- Erken, E.; Ozturk, O.G.; Kudas, O.; Tas, D.A.; Demirtas, A.; Kibar, F.; Dinkci, S.; Erken, E. Killer cell immunoglobulin-like receptor (KIR) genotype distribution in familial mediterranean fever (FMF) patients. Med. Sci. Monit. 2015, 21, 3547–3554. [Google Scholar] [CrossRef] [Green Version]
- Brusilovsky, M.; Rosental, B.; Shemesh, A.; Appel, M.Y.; Porgador, A. Human NK cell recognition of target cells in the prism of natural cytotoxicity receptors and their ligands. J. Immunotoxicol. 2012, 9, 267–274. [Google Scholar] [CrossRef] [Green Version]
- Vitale, M.; Cantoni, C.; Della Chiesa, M.; Ferlazzo, G.; Carlomagno, S.; Pende, D.; Falco, M.; Pessino, A.; Muccio, L.; De Maria, A.; et al. An Historical Overview: The Discovery of How NK Cells Can Kill Enemies, Recruit Defense Troops, and More. Front. Immunol. 2019, 10, 1415. [Google Scholar] [CrossRef] [Green Version]
- Ochoa, M.C.; Minute, L.; Rodriguez, I.; Garasa, S.; Perez-Ruiz, E.; Inogés, S.; Melero, I.; Berraondo, P. Antibody-dependent cell cytotoxicity: Immunotherapy strategies enhancing effector NK cells. Immunol. Cell Biol. 2017, 95, 347–355. [Google Scholar] [CrossRef] [PubMed]
- Sanchez-Correa, B.; Valhondo, I.; Hassouneh, F.; Lopez-Sejas, N.; Pera, A.; Bergua, J.M.; Arcos, M.J.; Bañas, H.; Casas-Avilés, I.; Durán, E.; et al. DNAM-1 and the TIGIT/PVRIG/TACTILE Axis: Novel Immune Checkpoints for Natural Killer Cell-Based Cancer Immunotherapy. Cancers 2019, 11, 877. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Petrie, E.J.; Clements, C.S.; Lin, J.; Sullivan, L.C.; Johnson, D.; Huyton, T.; Heroux, A.; Hoare, H.L.; Beddoe, T.; Reid, H.H.; et al. CD94-NKG2A recognition of human leukocyte antigen (HLA)-E bound to an HLA class I leader sequence. J. Exp. Med. 2008, 205, 725–735. [Google Scholar] [CrossRef] [Green Version]
- Borrego, F.; Masilamani, M.; Marusina, A.I.; Tang, X.; Coligan, J.E. The CD94/NKG2 family of receptors: From molecules and cells to clinical relevance. Immunol. Res. 2006, 35, 263–277. [Google Scholar] [CrossRef]
- Li, P.; Morris, D.L.; Willcox, B.E.; Steinle, A.; Spies, T.; Strong, R.K. Complex structure of the activating immunoreceptor NKG2D and its MHC class I-like ligand MICA. Nat. Immunol. 2001, 2, 443–451. [Google Scholar] [CrossRef] [PubMed]
- Pende, D.; Falco, M.; Vitale, M.; Cantoni, C.; Vitale, C.; Munari, E.; Bertaina, A.; Moretta, F.; Del Zotto, G.; Pietra, G.; et al. Killer Ig-Like Receptors (KIRs): Their Role in NK Cell Modulation and Developments Leading to Their Clinical Exploitation. Front. Immunol. 2019, 10, 1179. [Google Scholar] [CrossRef] [Green Version]
- Veillette, A. NK cell regulation by SLAM family receptors and SAP-related adapters. Immunol. Rev. 2006, 214, 22–34. [Google Scholar] [CrossRef] [PubMed]
- Khan, M.; Arooj, S.; Wang, H. NK Cell-Based Immune Checkpoint Inhibition. Front. Immunol. 2020, 11, 167. [Google Scholar] [CrossRef] [PubMed]
- Harjunpää, H.; Guillerey, C. TIGIT as an emerging immune checkpoint. Clin. Exp. Immunol. 2020, 200, 108–119. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Reches, A.; Ophir, Y.; Stein, N.; Kol, I.; Isaacson, B.; Charpak Amikam, Y.; Elnekave, A.; Tsukerman, P.; Kucan Brlic, P.; Lenac, T.; et al. Nectin4 is a novel TIGIT ligand which combines checkpoint inhibition and tumor specificity. J. Immunother. Cancer 2020, 8, e000266. [Google Scholar] [CrossRef]
- Chocarro, L.; Blanco, E.; Zuazo, M.; Arasanz, H.; Bocanegra, A.; Fernández-Rubio, L.; Morente, P.; Fernández-Hinojal, G.; Echaide, M.; Garnica, M.; et al. Understanding LAG-3 Signaling. Int. J. Mol. Sci. 2021, 22, 5282. [Google Scholar] [CrossRef] [PubMed]
- Wolf, Y.; Anderson, A.C.; Kuchroo, V.K. TIM3 comes of age as an inhibitory receptor. Nat. Rev. Immunol. 2020, 20, 173–185. [Google Scholar] [CrossRef] [PubMed]
- Georgiev, H.; Ravens, I.; Papadogianni, G.; Bernhardt, G. Coming of Age: CD96 Emerges as Modulator of Immune Responses. Front. Immunol. 2018, 9, 1072. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Noh, J.Y.; Yoon, S.R.; Kim, T.D.; Choi, I.; Jung, H. Toll-Like Receptors in Natural Killer Cells and Their Application for Immunotherapy. J. Immunol. Res. 2020, 2020. [Google Scholar] [CrossRef] [PubMed]
- O’Connor, G.M.; Hart, O.M.; Gardiner, C.M. Putting the natural killer cell in its place. Immunology 2005, 117, 1–10. [Google Scholar] [CrossRef]
- Iguchi-Manaka, A.; Kai, H.; Yamashita, Y.; Shibata, K.; Tahara-Hanaoka, S.; Honda, S.; Yasui, T.; Kikutani, H.; Shibuya, K.; Shibuya, A. Accelerated tumor growth in mice deficient in DNAM-1 receptor. J. Exp. Med. 2008, 205, 2959–2964. [Google Scholar] [CrossRef] [Green Version]
- Fuchs, A.; Cella, M.; Giurisato, E.; Shaw, A.S.; Colonna, M. Cutting edge: CD96 (tactile) promotes NK cell-target cell adhesion by interacting with the poliovirus receptor (CD155). J. Immunol. 2004, 172, 3994–3998. [Google Scholar] [CrossRef] [Green Version]
- Chiang, E.Y.; de Almeida, P.E.; de Almeida Nagata, D.E.; Bowles, K.H.; Du, X.; Chitre, A.S.; Banta, K.L.; Kwon, Y.; McKenzie, B.; Mittman, S.; et al. CD96 functions as a co-stimulatory receptor to enhance CD8(+) T cell activation and effector responses. Eur. J. Immunol. 2020, 50, 891–902. [Google Scholar] [CrossRef]
- Chan, C.J.; Martinet, L.; Gilfillan, S.; Souza-Fonseca-Guimaraes, F.; Chow, M.T.; Town, L.; Ritchie, D.S.; Colonna, M.; Andrews, D.M.; Smyth, M.J. The receptors CD96 and CD226 oppose each other in the regulation of natural killer cell functions. Nat. Immunol. 2014, 15, 431–438. [Google Scholar] [CrossRef]
- Blake, S.J.; Stannard, K.; Liu, J.; Allen, S.; Yong, M.C.R.; Mittal, D.; Aguilera, A.R.; Miles, J.J.; Lutzky, V.P.; de Andrade, L.F.; et al. Suppression of Metastases Using a New Lymphocyte Checkpoint Target for Cancer Immunotherapy. Cancer Discov. 2016, 6, 446–459. [Google Scholar] [CrossRef] [Green Version]
- Maghazachi, A.A. On the role of natural killer cells in neurodegenerative diseases. Toxins 2013, 5, 363–375. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Kadri, N.; Wagner, A.K.; Ganesan, S.; Kärre, K.; Wickström, S.; Johansson, M.H.; Höglund, P. Dynamic regulation of NK cell responsiveness. Curr. Top. Microbiol. Immunol. 2016, 395, 95–114. [Google Scholar] [CrossRef]
- Freud, A.G.; Mundy-Bosse, B.L.; Yu, J.; Caligiuri, M.A. The broad spectrum of human natural killer cell diversity. Immunity 2017, 47, 820–833. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Cooley, S.; Xiao, F.; Pitt, M.; Gleason, M.; McCullar, V.; Bergemann, T.; McQueen, K.; Guethlein, L.; Parham, P.; Miller, J. A subpopulation of human peripheral blood NK cells that lacks inhibitory receptors for self-MHC is developmentally immature. Blood 2007, 110, 578–586. [Google Scholar] [CrossRef] [Green Version]
- Fernandez, N.; Treiner, E.; Vance, R.; Jamieson, A.; Lemieux, S.; Raulet, D. A subset of natural killer cells achieves self-tolerance without expressing inhibitory receptors specific for self-MHC molecules. Blood 2005, 105, 4416–4423. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Kim, S.; Poursine-Laurent, J.; Truscott, S.; Lybarger, L.; Song, Y.; Yang, L.; French, A.; Sunwoo, J.; Lemieux, S.; Hansen, T.; et al. Licensing of natural killer cells by host major histocompatibility complex class I molecules. Nature 2005, 436, 709–713. [Google Scholar] [CrossRef] [PubMed]
- Anfossi, N.; André, P.; Guia, S.; Falk, C.; Roetynck, S.; Stewart, C.; Breso, V.; Frassati, C.; Reviron, D.; Middleton, D.; et al. Human NK cell education by inhibitory receptors for MHC class I. Immunity 2006, 25, 331–342. [Google Scholar] [CrossRef] [PubMed]
- Sordo-Bahamonde, C.; Lorenzo-Herrero, S.; Payer, Á.R.; Gonzalez, S.; López-Soto, A. Mechanisms of apoptosis resistance to NK cell-mediated cytotoxicity in cancer. Int. J. Mol. Sci. 2020, 21, 3726. [Google Scholar] [CrossRef]
- Lee, J.; Dieckmann, N.M.G.; Edgar, J.R.; Griffiths, G.M.; Siegel, R.M. Fas Ligand localizes to intraluminal vesicles within NK cell cytolytic granules and is enriched at the immune synapse. Immun. Inflamm. Dis. 2018, 6, 312–321. [Google Scholar] [CrossRef]
- Bossi, G.; Griffiths, G.M. Degranulation plays an essential part in regulating cell surface expression of Fas ligand in T cells and natural killer cells. Nat. Med. 1999, 5, 90–96. [Google Scholar] [CrossRef]
- Clavijo-Salomon, M.A.; Salcedo, R.; Roy, S.; Das Neves, R.X.; Dzutsev, A.; Sales-Campos, H.; Borbely, K.S.C.; Silla, L.; Orange, J.S.; Mace, E.M.; et al. Human NK cells prime inflammatory DC precursors to induce Tc17 differentiation. Blood Adv. 2020, 4, 3990–4006. [Google Scholar] [CrossRef] [PubMed]
- Sojka, D.K.; Yang, L.; Yokoyama, W.M. Uterine natural killer cells. Front. Immunol. 2019, 10, 960. [Google Scholar] [CrossRef] [PubMed]
- Vandenhaute, J.; Wouters, C.H.; Matthys, P. Natural Killer Cells in Systemic Autoinflammatory Diseases: A Focus on Systemic Juvenile Idiopathic Arthritis and Macrophage Activation Syndrome. Front. Immunol. 2020, 10, 3089. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Orange, J.S. Natural killer cell deficiency. J. Allergy Clin. Immunol. 2013, 132, 515–525. [Google Scholar] [CrossRef] [Green Version]
- Jiang, S.; Gao, H.; Luo, Q.; Wang, P.; Yang, X. The correlation of lymphocyte subsets, natural killer cell, and Parkinson’s disease: A meta-analysis. Neurol. Sci. 2017, 38, 1373–1380. [Google Scholar] [CrossRef] [PubMed]
- Poggi, A.; Zocchi, M.R. NK cell autoreactivity and autoimmune diseases. Front. Immunol. 2014, 5, 27. [Google Scholar] [CrossRef]
- Portales-Cervantes, L.; Dawod, B.; Marshall, J.S. Mast cells and natural killer cells—A potentially critical interaction. Viruses 2019, 11, 514. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Korbel, D.S.; Finney, O.C.; Riley, E.M. Natural killer cells and innate immunity to protozoan pathogens. Int. J. Parasitol. 2004, 34, 1517–1528. [Google Scholar] [CrossRef]
- Jost, S.; Altfeld, M. Control of human viral infections by natural killer cells. Annu. Rev. Immunol. 2013, 31, 163–194. [Google Scholar] [CrossRef]
- Ortaldo, J.R.; Wiltrout, R.H.; Reynolds, C.W. Natural killer activity: Early days, advances, and seminal observations. Crit. Rev. Oncog. 2014, 19, 1–13. [Google Scholar] [CrossRef] [PubMed]
- López-Soto, A.; Gonzalez, S.; Smyth, M.J.; Galluzzi, L. Control of Metastasis by NK Cells. Cancer Cell 2017, 32, 135–154. [Google Scholar] [CrossRef] [PubMed]
- Malmberg, K.J.; Carlsten, M.; Björklund, A.; Sohlberg, E.; Bryceson, Y.T.; Ljunggren, H.G. Natural killer cell-mediated immunosurveillance of human cancer. Semin. Immunol. 2017, 31, 20–29. [Google Scholar] [CrossRef]
- Shankaran, V.; Ikeda, H.; Bruce, A.T.; White, J.M.; Swanson, P.E.; Old, L.J.; Schreiber, R.D. IFNγ, and lymphocytes prevent primary tumour development and shape tumour immunogenicity. Nature 2001, 410, 1107–1111. [Google Scholar] [CrossRef] [PubMed]
- Street, S.; Cretney, E.; Smyth, M. Perforin and interferon-gamma activities independently control tumor initiation, growth, and metastasis. Blood 2001, 97, 192–197. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Van den Broek, M.; Kägi, D.; Ossendorp, F.; Toes, R.; Vamvakas, S.; Lutz, W.; Melief, C.; Zinkernagel, R.; Hengartner, H. Decreased tumor surveillance in perforin-deficient mice. J. Exp. Med. 1996, 184, 1781–1790. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Kaplan, D.; Shankaran, V.; Dighe, A.; Stockert, E.; Aguet, M.; Old, L.; Schreiber, R. Demonstration of an interferon gamma-dependent tumor surveillance system in immunocompetent mice. Proc. Natl. Acad. Sci. USA 1998, 95, 7556–7561. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Cretney, E.; Takeda, K.; Yagita, H.; Glaccum, M.; Peschon, J.J.; Smyth, M.J. Increased Susceptibility to Tumor Initiation and Metastasis in TNF-Related Apoptosis-Inducing Ligand-Deficient Mice. J. Immunol. 2002, 168, 1356–1361. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Takeda, K.; Smyth, M.; Cretney, E.; Hayakawa, Y.; Kayagaki, N.; Yagita, H.; Okumura, K. Critical role for tumor necrosis factor-related apoptosis-inducing ligand in immune surveillance against tumor development. J. Exp. Med. 2002, 195, 161–169. [Google Scholar] [CrossRef] [Green Version]
- Smyth, M.; Crowe, N.; Godfrey, D. NK cells and NKT cells collaborate in host protection from methylcholanthrene-induced fibrosarcoma. Int. Immunol. 2001, 13, 459–463. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Carlsten, M.; Malmberg, K.J.; Ljunggren, H.G. Natural killer cell-mediated lysis of freshly isolated human tumor cells. Int. J. Cancer 2009, 124, 757–762. [Google Scholar] [CrossRef] [PubMed]
- 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 2000, 356, 1795–1799. [Google Scholar] [CrossRef]
- Jouanguy, E.; Gineau, L.; Cottineau, J.; Béziat, V.; Vivier, E.; Casanova, J.L. Inborn errors of the development of human natural killer cells. Curr. Opin. Allergy Clin. Immunol. 2013, 13, 589–595. [Google Scholar] [CrossRef] [PubMed]
- Ebbo, M.; Gérard, L.; Carpentier, S.; Vély, F.; Cypowyj, S.; Farnarier, C.; Vince, N.; Malphettes, M.; Fieschi, C.; Oksenhendler, E.; et al. Low Circulating Natural Killer Cell Counts are Associated with Severe Disease in Patients with Common Variable Immunodeficiency. EBioMedicine 2016, 6, 222–230. [Google Scholar] [CrossRef] [Green Version]
- Hahn, C.N.; Chong, C.E.; Carmichael, C.L.; Wilkins, E.J.; Brautigan, P.J.; Li, X.C.; Babic, M.; Lin, M.; Carmagnac, A.; Lee, Y.K.; et al. Heritable GATA2 mutations associated with familial myelodysplastic syndrome and acute myeloid leukemia. Nat. Genet. 2011, 43, 1012–1017. [Google Scholar] [CrossRef]
- Shima, N.; Alcaraz, A.; Liachko, I.; Buske, T.R.; Andrews, C.A.; Munroe, R.J.; Hartford, S.A.; Tye, B.K.; Schimenti, J.C. A viable allele of Mcm4 causes chromosome instability and mammary adenocarcinomas in mice. Nat. Genet. 2007, 39, 93–98. [Google Scholar] [CrossRef] [PubMed]
- Jhunjhunwala, S.; Hammer, C.; Delamarre, L. Antigen presentation in cancer: Insights into tumour immunogenicity and immune evasion. Nat. Rev. Cancer 2021, 21, 298–312. [Google Scholar] [CrossRef]
- Jobim, M.; Jobim, L. Natural killer cells and immune surveillance. J. Pediatr. 2008, 84, S58–S67. [Google Scholar] [CrossRef] [Green Version]
- Huntington, N.; Cursons, J.; Rautela, J. The cancer-natural killer cell immunity cycle. Nat. Rev. Cancer 2020, 20, 437–454. [Google Scholar] [CrossRef] [PubMed]
- Smyth, M.; Godfrey, D.; Trapani, J. A fresh look at tumor immunosurveillance and immunotherapy. Nat. Immunol. 2001, 2, 293–299. [Google Scholar] [CrossRef]
- Sanchez-Martinez, D.; Allende-Vega, N.; Orecchioni, S.; Talarico, G.; Cornillon, A.; Vo, D.N.; Rene, C.; Lu, Z.Y.; Krzywinska, E.; Anel, A.; et al. Expansion of allogeneic NK cells with efficient antibody-dependent cell cytotoxicity against multiple tumors. Theranostics 2018, 8, 3856–3869. [Google Scholar] [CrossRef]
- Lequeux, A.; Noman, M.Z.; Xiao, M.; Van Moer, K.; Hasmim, M.; Benoit, A.; Bosseler, M.; Viry, E.; Arakelian, T.; Berchem, G.; et al. Targeting HIF-1 alpha transcriptional activity drives cytotoxic immune effector cells into melanoma and improves combination immunotherapy. Oncogene 2021, 40, 4725–4735. [Google Scholar] [CrossRef]
- Balsamo, M.; Manzini, C.; Pietra, G.; Raggi, F.; Blengio, F.; Mingari, M.C.; Varesio, L.; Moretta, L.; Bosco, M.C.; Vitale, M. Hypoxia downregulates the expression of activating receptors involved in NK-cell-mediated target cell killing without affecting ADCC. Eur. J. Immunol. 2013, 43, 2756–2764. [Google Scholar] [CrossRef] [PubMed]
- Prager, I.; Liesche, C.; van Ooijen, H.; Urlaub, D.; Verron, Q.; Sandström, N.; Fasbender, F.; Claus, M.; Eils, R.; Beaudouin, J.; et al. NK cells switch from granzyme B to death receptor–mediated cytotoxicity during serial killing. J. Exp. Med. 2019, 216, 2113–2127. [Google Scholar] [CrossRef] [Green Version]
- Rossin, A.; Miloro, G.; Hueber, A.O. TRAIL and FasL functions in cancer and autoimmune diseases: Towards an increasing complexity. Cancers 2019, 11, 639. [Google Scholar] [CrossRef] [Green Version]
- Bhat, R.; Watzl, C. Serial Killing of Tumor Cells by Human Natural Killer Cells—Enhancement by Therapeutic Antibodies. PLoS ONE 2007, 2, e326. [Google Scholar] [CrossRef]
- Dunn, G.; Bruce, A.; Ikeda, H.; Old, L.; Schreiber, R. Cancer immunoediting: From immunosurveillance to tumor escape. Nat. Immunol. 2002, 3, 991–998. [Google Scholar] [CrossRef]
- Zlotnik, A.; Yoshie, O. The chemokine superfamily revisited. Immunity 2012, 36, 705–716. [Google Scholar] [CrossRef] [Green Version]
- Shida, Y.; Nakazawa, T.; Matsuda, R.; Morimoto, T.; Nishimura, F.; Nakamura, M.; Maeoka, R.; Yamada, S.; Nakagawa, I.; Park, Y.-S.; et al. Ex Vivo Expanded and Activated Natural Killer Cells Prolong the Overall Survival of Mice with Glioblastoma-like Cell-Derived Tumors. Int. J. Mol. Sci. 2021, 22, 9975. [Google Scholar] [CrossRef]
- Maghazachi, A.A. Role of Chemokines in the Biology of Natural Killer Cells. In Current Topics in Microbiology and Immunology; Springer: Berlin/Heidelberg, Germany, 2010; Volume 341, pp. 37–58. [Google Scholar]
- Schall, T.J.; Bacon, K.; Toy, K.J.; Goeddel, D. V Selective attraction of monocytes and T lymphocytes of the memory phenotype by cytokine RANTES. Nature 1990, 347, 669–671. [Google Scholar] [CrossRef] [PubMed]
- Maghazachi, A.A.; Al-Aoukaty, A.; Schall, T.J. CC chemokines induce the generation of killer cells from CD56+ cells. Eur. J. Immunol. 1996, 26, 315–319. [Google Scholar] [CrossRef]
- Martín-Fontecha, A.; Thomsen, L.; Brett, S.; Gerard, C.; Lipp, M.; Lanzavecchia, A.; Sallusto, F. Induced recruitment of NK cells to lymph nodes provides IFN-gamma for T(H)1 priming. Nat. Immunol. 2004, 5, 1260–1265. [Google Scholar] [CrossRef] [PubMed]
- Böttcher, J.P.; Bonavita, E.; Chakravarty, P.; Blees, H.; Cabeza-Cabrerizo, M.; Sammicheli, S.; Rogers, N.C.; Sahai, E.; Zelenay, S.; Reis e Sousa, C. NK Cells Stimulate Recruitment of cDC1 into the Tumor Microenvironment Promoting Cancer Immune Control. Cell 2018, 172, 1022–1037. [Google Scholar] [CrossRef] [Green Version]
- Barry, K.C.; Hsu, J.; Broz, M.L.; Cueto, F.J.; Binnewies, M.; Combes, A.J.; Nelson, A.E.; Loo, K.; Kumar, R.; Rosenblum, M.D.; et al. A natural killer-dendritic cell axis defines checkpoint therapy-responsive tumor microenvironments. Nat. Med. 2018, 24, 1178–1191. [Google Scholar] [CrossRef]
- Sánchez-Paulete, A.R.; Cueto, F.J.; Martínez-López, M.; Labiano, S.; Morales-Kastresana, A.; Rodríguez-Ruiz, M.E.; Jure-Kunkel, M.; Azpilikueta, A.; Aznar, M.A.; Quetglas, J.I.; et al. Cancer immunotherapy with immunomodulatory anti-CD137 and anti-PD-1 monoclonal antibodies requires Batf3-dependent dendritic cells. Cancer Discov. 2016, 6, 71–79. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Hildner, K.; Edelson, B.; Purtha, W.; Diamond, M.; Matsushita, H.; Kohyama, M.; Calderon, B.; Schraml, B.; Unanue, E.; Diamond, M.; et al. Batf3 deficiency reveals a critical role for CD8alpha+ dendritic cells in cytotoxic T cell immunity. Science 2008, 322, 1097–1100. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Deauvieau, F.; Ollion, V.; Doffin, A.C.; Achard, C.; Fonteneau, J.F.; Verronese, E.; Durand, I.; Ghittoni, R.; Marvel, J.; Dezutter-Dambuyant, C.; et al. Human natural killer cells promote cross-presentation of tumor cell-derived antigens by dendritic cells. Int. J. Cancer 2015, 136, 1085–1094. [Google Scholar] [CrossRef]
- Mocikat, R.; Braumüller, H.; Gumy, A.; Egeter, O.; Ziegler, H.; Reusch, U.; Bubeck, A.; Louis, J.; Mailhammer, R.; Riethmüller, G.; et al. Natural killer cells activated by MHC class I(low) targets prime dendritic cells to induce protective CD8 T cell responses. Immunity 2003, 19, 561–569. [Google Scholar] [CrossRef] [Green Version]
- Kelly, J.; Darcy, P.; Markby, J.; Godfrey, D.; Takeda, K.; Yagita, H.; Smyth, M. Induction of tumor-specific T cell memory by NK cell-mediated tumor rejection. Nat. Immunol. 2002, 3, 83–90. [Google Scholar] [CrossRef]
- Nersesian, S.; Schwartz, S.L.; Grantham, S.R.; MacLean, L.K.; Lee, S.N.; Pugh-Toole, M.; Boudreau, J.E. NK cell infiltration is associated with improved overall survival in solid cancers: A systematic review and meta-analysis. Transl. Oncol. 2021, 14, 100930. [Google Scholar] [CrossRef] [PubMed]
- Galea, I.; Bechmann, I.; Perry, V.H. What is immune privilege (not)? Trends Immunol. 2007, 28, 12–18. [Google Scholar] [CrossRef] [PubMed]
- Watkins, S.; Robel, S.; Kimbrough, I.F.; Robert, S.M.; Ellis-Davies, G.; Sontheimer, H. Disruption of astrocyte-vascular coupling and the blood-brain barrier by invading glioma cells. Nat. Commun. 2014, 5, 4196. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Orrego, E.; Castaneda, C.A.; Castillo, M.; Bernabe, L.A.; Casavilca, S.; Chakravarti, A.; Meng, W.; Garcia-Corrochano, P.; Villa-Robles, M.R.; Zevallos, R.; et al. Distribution of tumor-infiltrating immune cells in glioblastoma. CNS Oncol. 2018, 7, CNS21. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Yang, I.; Han, S.J.; Sughrue, M.E.; Tihan, T.; Parsa, A.T. Immune cell infiltrate differences in pilocytic astrocytoma and glioblastoma: Evidence of distinct immunological microenvironments that reflect tumor biology. J. Neurosurg. 2011, 115, 505–511. [Google Scholar] [CrossRef]
- Kmiecik, J.; Poli, A.; Brons, N.H.C.; Waha, A.; Eide, G.E.; Enger, P.Ø.; Zimmer, J.; Chekenya, M. Elevated CD3+ and CD8+ tumor-infiltrating immune cells correlate with prolonged survival in glioblastoma patients despite integrated immunosuppressive mechanisms in the tumor microenvironment and at the systemic level. J. Neuroimmunol. 2013, 264, 71–83. [Google Scholar] [CrossRef] [Green Version]
- González-Tablas Pimenta, M.; Otero, Á.; Arandia Guzman, D.A.; Pascual-Argente, D.; Ruíz Martín, L.; Sousa-Casasnovas, P.; García-Martin, A.; de Oca Roa Montes, J.C.; Villaseñor-Ledezma, J.; Torres Carretero, L.; et al. Tumor cell and immune cell profiles in primary human glioblastoma: Impact on patient outcome. Brain Pathol. 2021, 31, 365–380. [Google Scholar] [CrossRef]
- Tan, W.C.C.; Nerurkar, S.N.; Cai, H.Y.; Ng, H.H.M.; Wu, D.; Wee, Y.T.F.; Lim, J.C.T.; Yeong, J.; Lim, T.K.H. Overview of multiplex immunohistochemistry/immunofluorescence techniques in the era of cancer immunotherapy. Cancer Commun. 2020, 40, 135–153. [Google Scholar] [CrossRef] [Green Version]
- Chen, B.; Khodadoust, M.S.; Liu, C.L.; Newman, A.M.; Alizadeh, A.A. Profiling tumor infiltrating immune cells with CIBERSORT. In Cancer Systems Biology. Methods and Protocols; Von Stechow, L., Ed.; Humana Press Inc.: New York, NY, USA, 2018; Volume 1711, pp. 243–259. ISBN 978-1-4939-7492-4. [Google Scholar]
- Wang, J.; Matosevic, S. NT5E/CD73 as Correlative Factor of Patient Survival and Natural Killer Cell Infiltration in Glioblastoma. J. Clin. Med. 2019, 8, 1526. [Google Scholar] [CrossRef] [Green Version]
- Close, H.J.; Stead, L.F.; Nsengimana, J.; Reilly, K.A.; Droop, A.; Wurdak, H.; Mathew, R.K.; Corns, R.; Newton-Bishop, J.; Melcher, A.A.; et al. Expression profiling of single cells and patient cohorts identifies multiple immunosuppressive pathways and an altered NK cell phenotype in glioblastoma. Clin. Exp. Immunol. 2020, 200, 33–44. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Fu, W.; Wenjing, W.; Hao, L.; Yuming, J.; Huo, R.; Zihan, Y.; Wang, J.; Wang, S.; Wang, J.; Chen, D.; et al. Single-Cell Atlas Reveals Complexity of the Immunosuppressive Microenvironment of Initial and Recurrent Glioblastoma. Front. Immunol. 2020, 11, 835. [Google Scholar] [CrossRef] [PubMed]
- Romee, R.; Foley, B.; Lenvik, T.; Wang, Y.; Zhang, B.; Ankarlo, D.; Luo, X.; Cooley, S.; Verneris, M.; Walcheck, B.; et al. NK cell CD16 surface expression and function is regulated by a disintegrin and metalloprotease-17 (ADAM17). Blood 2013, 121, 3599–3608. [Google Scholar] [CrossRef]
- Grzywacz, B.; Kataria, N.; Verneris, M.R. CD56dimCD16+ NK cells downregulate CD16 following target cell induced activation of matrix metalloproteinases. Leukemia 2007, 21, 356–359. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Vujanovic, L.; Chuckran, C.; Lin, Y.; Ding, F.; Sander, C.A.; Santos, P.M.; Lohr, J.; Mashadi-Hossein, A.; Warren, S.; White, A.; et al. CD56dim CD16− Natural Killer Cell Profiling in Melanoma Patients Receiving a Cancer Vaccine and Interferon-α. Front. Immunol. 2019, 10, 14. [Google Scholar] [CrossRef] [PubMed]
- Wendel, M.; Galani, I.E.; Suri-Payer, E.; Cerwenka, A. Natural killer cell accumulation in tumors is dependent on IFN-γ and CXCR3 ligands. Cancer Res. 2008, 68, 8437–8445. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Lee, S.J.; Kang, W.Y.; Yoon, Y.; Jin, J.Y.; Song, H.J.; Her, J.H.; Kang, S.M.; Hwang, Y.K.; Kang, K.J.; Joo, K.M.; et al. Natural killer (NK) cells inhibit systemic metastasis of glioblastoma cells and have therapeutic effects against glioblastomas in the brain. BMC Cancer 2015, 15, 1011. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Beauchesne, P. Extra-neural metastases of malignant gliomas: Myth or reality? Cancers 2011, 3, 461–477. [Google Scholar] [CrossRef] [Green Version]
- Stevens, A.; Klöter, I.; Roggendorf, W. Inflammatory infiltrates and natural killer cell presence in human brain tumors. Cancer 1988, 61, 738–743. [Google Scholar] [CrossRef]
- Vaquero, J.; Coca, S.; Oya, S.; Martínez, R.; Ramiro, J.; Salazar, F. Presence and significance of NK cells in glioblastomas. J. Neurosurg. 1989, 70, 728–731. [Google Scholar] [CrossRef]
- Liang, P.; Chai, Y.; Zhao, H.; Wang, G. Predictive Analyses of Prognostic-Related Immune Genes and Immune Infiltrates for Glioblastoma. Diagnostics 2020, 10, 177. [Google Scholar] [CrossRef] [Green Version]
- Mostafa, H.; Pala, A.; Högel, J.; Hlavac, M.; Dietrich, E.; Westhoff, M.; Nonnenmacher, L.; Burster, T.; Georgieff, M.; Wirtz, C.; et al. Immune phenotypes predict survival in patients with glioblastoma multiforme. J. Hematol. Oncol. 2016, 9, 77. [Google Scholar] [CrossRef] [Green Version]
- Kmiecik, J.; Zimmer, J.; Chekenya, M. Natural killer cells in intracranial neoplasms: Presence and therapeutic efficacy against brain tumours. J. Neurooncol. 2014, 116, 1–9. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Razavi, S.-M.; Lee, K.E.; Jin, B.E.; Aujla, P.S.; Gholamin, S.; Li, G. Immune Evasion Strategies of Glioblastoma. Front. Surg. 2016, 3, 11. [Google Scholar] [CrossRef]
- Roth, P.; Mittelbronn, M.; Wick, W.; Meyermann, R.; Tatagiba, M.; Weller, M. Malignant glioma cells counteract antitumor immune responses through expression of lectin-like transcript-1. Cancer Res. 2007, 67, 3540–3544. [Google Scholar] [CrossRef] [Green Version]
- Wischhusen, J.; Friese, M.; Mittelbronn, M.; Meyermann, R.; Weller, M. HLA-E protects glioma cells from NKG2D-mediated immune responses in vitro: Implications for immune escape in vivo. J. Neuropathol. Exp. Neurol. 2005, 64, 523–528. [Google Scholar] [CrossRef] [PubMed]
- Zhang, X.; Wu, A.; Fan, Y.; Wang, Y. Increased transforming growth factor-β2 in epidermal growth factor receptor variant III-positive glioblastoma. J. Clin. Neurosci. 2011, 18, 821–826. [Google Scholar] [CrossRef]
- Zwirner, N.W.; Domaica, C.I. Cytokine regulation of natural killer cell effector functions. BioFactors 2010, 36, 274–288. [Google Scholar] [CrossRef] [PubMed]
- Castriconi, R.; Cantoni, C.; Della Chiesa, M.; Vitale, M.; Marcenaro, E.; Conte, R.; Biassoni, R.; Bottino, C.; Moretta, L.; Moretta, A. Transforming growth factor beta 1 inhibits expression of NKp30 and NKG2D receptors: Consequences for the NK-mediated killing of dendritic cells. Proc. Natl. Acad. Sci. USA 2003, 100, 4120–4125. [Google Scholar] [CrossRef] [Green Version]
- Wilson, E.B.; El-Jawhari, J.J.; Neilson, A.L.; Hall, G.D.; Melcher, A.A.; Meade, J.L.; Cook, G.P. Human tumour immune evasion via TGF-β blocks NK cell activation but not survival allowing therapeutic restoration of anti-tumour activity. PLoS ONE 2011, 6, 22842. [Google Scholar] [CrossRef] [Green Version]
- Burnet, M. Cancer: A Biological Approach. III. Viruses associated with neoplastic conditions. IV.Pratical aplications. Br. Med. J. 1957, 1, 841–847. [Google Scholar] [CrossRef] [PubMed]
- Helmy, K.Y.; Patel, S.A.; Nahas, G.R.; Rameshwar, P. Cancer immunotherapy: Accomplishments to date and future promise. Ther. Deliv. 2013, 4, 1307–1320. [Google Scholar] [CrossRef]
- Choucair, K.; Duff, J.R.; Cassidy, C.S.; Albrethsen, M.T.; Kelso, J.D.; Lenhard, A.; Staats, H.; Patel, R.; Brunicardi, F.C.; Dworkin, L.; et al. Natural killer cells: A review of biology, therapeutic potential and challenges in treatment of solid tumors. Future Oncol. 2019, 15, 3053–3069. [Google Scholar] [CrossRef]
- Shimasaki, N.; Jain, A.; Campana, D. NK cells for cancer immunotherapy. Nat. Rev. Drug Discov. 2020, 19, 200–218. [Google Scholar] [CrossRef] [PubMed]
- Ruggeri, L.; Capanni, M.; Urbani, E.; Perruccio, K.; Shlomchik, W.D.; Tosti, A.; Posati, S.; Rogaia, D.; Frassoni, F.; Aversa, F.; et al. Effectiveness of donor natural killer cell alloreactivity in mismatched hematopoietic transplants. Science 2002, 295, 2097–2100. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Miller, J.S.; Soignier, Y.; Panoskaltsis-Mortari, A.; McNearney, S.A.; Yun, G.H.; Fautsch, S.K.; McKenna, D.; Le, C.; Defor, T.E.; Burns, L.J.; et al. Successful adoptive transfer and in vivo expansion of human haploidentical NK cells in patients with cancer. Blood 2005, 105, 3051–3057. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Liu, E.; Marin, D.; Banerjee, P.; Macapinlac, H.A.; Thompson, P.; Basar, R.; Nassif Kerbauy, L.; Overman, B.; Thall, P.; Kaplan, M.; et al. Use of CAR-Transduced Natural Killer Cells in CD19-Positive Lymphoid Tumors. N. Engl. J. Med. 2020, 382, 545–553. [Google Scholar] [CrossRef]
- Fang, F.; Xiao, W.; Tian, Z. NK cell-based immunotherapy for cancer. Semin. Immunol. 2017, 31, 37–54. [Google Scholar] [CrossRef] [PubMed]
- Navarro, A.G.; Espedal, H.; Joseph, J.V.; Trachsel-Moncho, L.; Bahador, M.; Gjertsen, B.T.; Kristoffersen, E.K.; Simonsen, A.; Miletic, H.; Enger, P.Ø.; et al. Pretreatment of glioblastoma with bortezomib potentiates natural killer cell cytotoxicity through TRAIL/DR5 mediated apoptosis and prolongs animal survival. Cancers 2019, 11, 996. [Google Scholar] [CrossRef] [Green Version]
- Jung, T.-Y.; Choi, Y.-D.; Kim, Y.-H.; Lee, J.-J.; Kim, H.-S.; Kim, J.-S.; Kim, S.-K.; Jung, S.; Cho, D. Immunological characterization of glioblastoma cells for immunotherapy. Anticancer Res. 2013, 33, 2525–2533. [Google Scholar]
- Oh, S.J.; Yang, J.I.; Kim, O.; Ahn, E.J.; Kang, W.D.; Lee, J.H.; Moon, K.S.; Lee, K.H.; Cho, D. Human U87 glioblastoma cells with stemness features display enhanced sensitivity to natural killer cell cytotoxicity through altered expression of NKG2D ligand. Cancer Cell Int. 2017, 17, 22. [Google Scholar] [CrossRef] [Green Version]
- Sharifzad, F.; Mardpour, S.; Mardpour, S.; Fakharian, E.; Taghikhani, A.; Sharifzad, A.; Kiani, S.; Heydarian, Y.; Łos, M.; Azizi, Z.; et al. HSP70/IL-2 Treated NK Cells Effectively Cross the Blood Brain Barrier and Target Tumor Cells in a Rat Model of Induced Glioblastoma Multiforme (GBM). Int. J. Mol. Sci. 2020, 21, 2263. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Tanaka, Y.; Nakazawa, T.; Nakamura, M.; Nishimura, F.; Matsuda, R.; Omoto, K.; Shida, Y.; Murakami, T.; Nakagawa, I.; Motoyama, Y.; et al. Ex vivo-expanded highly purified natural killer cells in combination with temozolomide induce antitumor effects in human glioblastoma cells in vitro. PLoS ONE 2019, 14, e0212455. [Google Scholar] [CrossRef] [PubMed]
- Giannattasio, A.; Weil, S.; Kloess, S.; Ansari, N.; Stelzer, E.H.K.; Cerwenka, A.; Steinle, A.; Koehl, U.; Koch, J. Cytotoxicity and infiltration of human NK cells in in vivo-like tumor spheroids. BMC Cancer 2015, 15, 351. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Morimoto, T.; Nakazawa, T.; Matsuda, R.; Nishimura, F.; Nakamura, M.; Yamada, S.; Nakagawa, I.; Park, Y.-S.; Tsujimura, T.; Nakase, H. Evaluation of Comprehensive Gene Expression and NK Cell-Mediated Killing in Glioblastoma Cell Line-Derived Spheroids. Cancers 2021, 13, 4896. [Google Scholar] [CrossRef]
- Nunes, A.S.; Barros, A.S.; Costa, E.C.; Moreira, A.F.; Correia, I.J. 3D tumor spheroids as in vitro models to mimic in vivo human solid tumors resistance to therapeutic drugs. Biotechnol. Bioeng. 2019, 116, 206–226. [Google Scholar] [CrossRef] [Green Version]
- Haspels, H.N.; Rahman, M.A.; Joseph, J.V.; Navarro, A.G.; Chekenya, M. Glioblastoma Stem-Like Cells Are More Susceptible Than Differentiated Cells to Natural Killer Cell Lysis Mediated Through Killer Immunoglobulin-Like Receptors–Human Leukocyte Antigen Ligand Mismatch and Activation Receptor–Ligand Interactions. Front. Immunol. 2018, 9, 1345. [Google Scholar] [CrossRef]
- Avril, T.; Vauleon, E.; Hamlat, A.; Saikali, S.; Etcheverry, A.; Delmas, C.; Diabira, S.; Mosser, J.; Quillien, V. Human glioblastoma stem-like cells are more sensitive to allogeneic NK and T cell-mediated killing compared with serum-cultured glioblastoma cells. Brain Pathol. 2012, 22, 159–174. [Google Scholar] [CrossRef] [PubMed]
- Castriconi, R.; Daga, A.; Dondero, A.; Zona, G.; Poliani, P.L.; Melotti, A.; Griffero, F.; Marubbi, D.; Spaziante, R.; Bellora, F.; et al. NK Cells Recognize and Kill Human Glioblastoma Cells with Stem Cell-Like Properties. J. Immunol. 2009, 182, 3530–3539. [Google Scholar] [CrossRef] [Green Version]
- Tseng, H.-C.; Inagaki, A.; Bui, V.T.; Cacalano, N.; Kasahara, N.; Man, Y.; Jewett, A. Differential Targeting of Stem Cells and Differentiated Glioblastomas by NK Cells. J. Cancer 2015, 6, 866–876. [Google Scholar] [CrossRef] [Green Version]
- Shaim, H.; Shanley, M.; Basar, R.; Daher, M.; Gumin, J.; Zamler, D.B.; Uprety, N.; Wang, F.; Huang, Y.; Gabrusiewicz, K.; et al. Targeting the αv integrin/TGF-β axis improves natural killer cell function against glioblastoma stem cells. J. Clin. Investig. 2021, 131. [Google Scholar] [CrossRef]
- Gimple, R.C.; Bhargava, S.; Dixit, D.; Rich, J.N. Glioblastoma stem cells: Lessons from the tumor hierarchy in a lethal cancer. Genes Dev. 2019, 33, 591–609. [Google Scholar] [CrossRef]
- Backström, E.; Chambers, B.J.; Ho, E.L.; Naidenko, O.V.; Mariotti, R.; Fremont, D.H.; Yokoyama, W.M.; Kristensson, K.; Ljunggren, H.-G. Natural killer cell-mediated lysis of dorsal root ganglia neurons via RAE1/NKG2D interactions. Eur. J. Immunol. 2003, 33, 92–100. [Google Scholar] [CrossRef]
- Davies, A.J.; Kim, H.W.; Gonzalez-Cano, R.; Choi, J.; Back, S.K.; Roh, S.E.; Johnson, E.; Gabriac, M.; Kim, M.-S.; Lee, J.; et al. Natural Killer Cells Degenerate Intact Sensory Afferents following Nerve Injury. Cell 2019, 176, 716–728.e18. [Google Scholar] [CrossRef] [Green Version]
- Backström, E.; Ljunggren, H.-G.; Kristensson, K. NK cell-mediated destruction of influenza A virus-infected peripheral but not central neurones. Scand. J. Immunol. 2007, 65, 353–361. [Google Scholar] [CrossRef] [PubMed]
- Backström, E.; Chambers, B.J.; Kristensson, K.; Ljunggren, H.G. Direct NK cell-mediated lysis of syngenic dorsal root ganglia neurons in vitro. J. Immunol. 2000, 165, 4895–4900. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Jin, W.-N.; Shi, K.; He, W.; Sun, J.-H.; Van Kaer, L.; Shi, F.-D.; Liu, Q. Neuroblast senescence in the aged brain augments natural killer cell cytotoxicity leading to impaired neurogenesis and cognition. Nat. Neurosci. 2021, 24, 61–73. [Google Scholar] [CrossRef]
- Nianias, A.; Themeli, M. Induced Pluripotent Stem Cell (iPSC)–Derived Lymphocytes for Adoptive Cell Immunotherapy: Recent Advances and Challenges. Curr. Hematol. Malig. Rep. 2019, 14, 261–268. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Oberoi, P.; Kamenjarin, K.; Ossa, J.; Uherek, B.; Bönig, H.; Wels, W. Directed Differentiation of Mobilized Hematopoietic Stem and Progenitor Cells into Functional NK cells with Enhanced Antitumor Activity. Cells 2020, 9, 811. [Google Scholar] [CrossRef] [Green Version]
- Suck, G.; Odendahl, M.; Nowakowska, P.; Seidl, C.; Wels, W.S.; Klingemann, H.G.; Tonn, T. NK-92: An ‘off-the-shelf therapeutic’ for adoptive natural killer cell-based cancer immunotherapy. Cancer Immunol. Immunother. 2015, 65, 485–492. [Google Scholar] [CrossRef] [PubMed]
- Daher, M.; Rezvani, K. Next generation natural killer cells for cancer immunotherapy: The promise of genetic engineering. Curr. Opin. Immunol. 2018, 51, 146–153. [Google Scholar] [CrossRef]
- Ishikawa, E.; Tsuboi, K.; Saijo, K.; Harada, H.; Takano, S.; Nose, T.; Ohno, T. Autologous natural killer cell therapy for human recurrent malignant glioma. Anticancer Res. 2004, 24, 1861–1871. [Google Scholar]
- Dillman, R.; Duma, C.; Schiltz, P.; DePriest, C.; Ellis, R.; Okamoto, K.; Beutel, L.; De Leon, C.; Chico, S. Intracavitary placement of autologous lymphokine-activated killer (LAK) cells after resection of recurrent glioblastoma. J. Immunother. 2004, 27, 398–404. [Google Scholar] [CrossRef]
- Sankhla, S.K.; Nadkarni, J.S.; Bhagwati, S.N. Adoptive immunotherapy using lymphokine-activated killer (LAK) cells and interleukin-2 for recurrent malignant primary brain tumors. J. Neurooncol. 1996, 27, 133–140. [Google Scholar] [CrossRef]
- Cao, J.-X.; Gao, W.-J.; You, J.; Wu, L.-H.; Wang, Z.-X. Assessment of the efficacy of passive cellular immunotherapy for glioma patients. Rev. Neurosci. 2020, 31, 427–440. [Google Scholar] [CrossRef]
- Parkhurst, M.; Riley, J.; Dudley, M.; Rosenberg, S. 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. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Nagai, K.; Harada, Y.; Harada, H.; Yanagihara, K.; Yonemitsu, Y.; Miyazaki, Y. Highly Activated Ex Vivo-expanded Natural Killer Cells in Patients with Solid Tumors in a Phase I/IIa Clinical Study. Anticancer Res. 2020, 40, 5687–5700. [Google Scholar] [CrossRef] [PubMed]
- Li, L.; Li, W.; Wang, C.; Yan, X.; Wang, Y.; Niu, C.; Zhang, X.; Li, M.; Tian, H.; Yao, C.; et al. Adoptive transfer of natural killer cells in combination with chemotherapy improves outcomes of patients with locally advanced colon carcinoma. Cytotherapy 2018, 20, 134–148. [Google Scholar] [CrossRef]
- Itoh, T.; Ueda, Y.; Okugawa, K.; Fujiwara, H.; Fuji, N.; Yamashita, T.; Fujiki, H.; Harada, S.; Yoshimura, T.; Yamagishi, H. Streptococcal preparation OK432 promotes functional maturation of human monocyte-derived dendritic cells. Cancer Immunol. Immunother. 2003, 52, 207–214. [Google Scholar] [CrossRef]
- Merchant, R.E.; Grant, A.J.; Merchant, L.H.; Young, H.F. Adoptive immunotherapy for recurrent glioblastoma multiforme using lymphokine activated killer cells and recombinant interleukin-2. Cancer 1988, 62, 665–671. [Google Scholar] [CrossRef]
- Blancher, A.; Roubinet, F.; Grancher, A.S.; Tremoulet, M.; Bonaté, A.; Delisle, M.B.; Calot, J.P.; Pourreau, C.; Franks, C.; Ducos, J. Local immunotherapy of recurrent glioblastoma multiforme by intracerebral perfusion of interleukin-2 and LAK cells. Eur. Cytokine Netw. 1993, 4, 331–341. [Google Scholar]
- Dillman, R.; Duma, C.; Ellis, R.; Cornforth, A.; Schiltz, P.; Sharp, S.; DePriest, M. Intralesional lymphokine-activated killer cells as adjuvant therapy for primary glioblastoma. J. Immunother. 2009, 32, 914–919. [Google Scholar] [CrossRef]
- Richards, J. Therapeutic uses of interleukin-2 and lymphokine-activated killer (LAK) cells. Blood Rev. 1989, 3, 110–119. [Google Scholar] [CrossRef]
- Ciocca, D.R.; Calderwood, S.K. Heat shock proteins in cancer: Diagnostic, prognostic, predictive, and treatment implications. Cell Stress Chaperones 2005, 10, 86–103. [Google Scholar] [CrossRef]
- Hermisson, M.; Strik, H.; Rieger, J.; Dichgans, J.; Meyermann, R.; Weller, M. Expression and functional activity of heat shock proteins in human glioblastoma multiforme. Neurology 2000, 54, 1357–1365. [Google Scholar] [CrossRef] [PubMed]
- Lobinger, D.; Gempt, J.; Sievert, W.; Barz, M.; Schmitt, S.; Nguyen, H.T.; Stangl, S.; Werner, C.; Wang, F.; Wu, Z.; et al. Potential Role of Hsp70 and Activated NK Cells for Prediction of Prognosis in Glioblastoma Patients. Front. Mol. Biosci. 2021, 8, 669366. [Google Scholar] [CrossRef]
- Krause, S.; Gastpar, R.; Andreesen, R.; Gross, C.; Ullrich, H.; Thonigs, G.; Pfister, K.; Multhoff, G. Treatment of colon and lung cancer patients with ex vivo heat shock protein 70-peptide-activated, autologous natural killer cells: A clinical phase i trial. Clin. Cancer Res. 2004, 10, 3699–3707. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Ruggeri, L.; Mancusi, A.; Capanni, M.; Martelli, M.; Velardi, A. Exploitation of alloreactive NK cells in adoptive immunotherapy of cancer. Curr. Opin. Immunol. 2005, 17, 211–217. [Google Scholar] [CrossRef] [PubMed]
- Yang, Y.; Lim, O.; Kim, T.M.; Ahn, Y.-O.; Choi, H.; Chung, H.; Min, B.; Her, J.H.; Cho, S.Y.; Keam, B.; et al. Phase I Study of Random Healthy Donor-Derived Allogeneic Natural Killer Cell Therapy in Patients with Malignant Lymphoma or Advanced Solid Tumors. Cancer Immunol. Res. 2016, 4, 215–224. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Wang, W.; Erbe, A.K.; DeSantes, K.B.; Sondel, P.M. Donor selection for ex vivo-expanded natural killer cells as adoptive cancer immunotherapy. Future Oncol. 2017, 13, 1043–1047. [Google Scholar] [CrossRef] [Green Version]
- Siegler, U.; Meyer-Monard, S.; Jörger, S.; Stern, M.; Tichelli, A.; Gratwohl, A.; Wodnar-Filipowicz, A.; Kalberer, C.P. Good manufacturing practice-compliant cell sorting and large-scale expansion of single KIR-positive alloreactive human natural killer cells for multiple infusions to leukemia patients. Cytotherapy 2010, 12, 750–763. [Google Scholar] [CrossRef]
- Natural Killer Cell (CYNK-001) IV Infusion or IT Administration in Adults with Recurrent GBM—ClinicalTrials.gov. Available online: https://clinicaltrials.gov/ct2/show/NCT04489420?cond=NCT04489420&draw=2&rank=1 (accessed on 10 August 2021).
- Eshhar, Z.; Waks, T.; Gross, G.; Schindler, D.G. Specific activation and targeting of cytotoxic lymphocytes through chimeric single chains consisting of antibody-binding domains and the gamma or zeta subunits of the immunoglobulin and T-cell receptors. Proc. Natl. Acad. Sci. USA 1993, 90, 720–724. [Google Scholar] [CrossRef] [Green Version]
- Guedan, S.; Calderon, H.; Posey, A.D.J.; Maus, M.V. Engineering and Design of Chimeric Antigen Receptors. Mol. Ther. Methods Clin. Dev. 2019, 12, 145–156. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Hunter, B.D.; Jacobson, C.A. CAR T-Cell Associated Neurotoxicity: Mechanisms, Clinicopathologic Correlates, and Future Directions. J. Natl. Cancer Inst. 2019, 111, 646–654. [Google Scholar] [CrossRef] [PubMed]
- Tang, X.; Yang, L.; Li, Z.; Nalin, A.P.; Dai, H.; Xu, T.; Yin, J.; You, F.; Zhu, M.; Shen, W.; 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. [Google Scholar] [PubMed]
- Karadimitris, A. Cord Blood CAR-NK Cells: Favorable Initial Efficacy and Toxicity but Durability of Clinical Responses Not Yet Clear. Cancer Cell 2020, 37, 426–427. [Google Scholar] [CrossRef] [PubMed]
- 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. 1998, 28, 780–786. [Google Scholar] [CrossRef]
- Pfefferle, A.; Huntington, N.D. You Have Got a Fast CAR: Chimeric Antigen Receptor NK Cells in Cancer Therapy. Cancers 2020, 12, 706. [Google Scholar] [CrossRef] [Green Version]
- Oberschmidt, O.; Kloess, S.; Koehl, U. Redirected Primary Human Chimeric Antigen Receptor Natural Killer Cells As an “Off-the-Shelf Immunotherapy” for Improvement in Cancer Treatment. Front. Immunol. 2017, 8, 654. [Google Scholar] [CrossRef] [Green Version]
- Marcus, A.; Eshhar, Z. Allogeneic chimeric antigen receptor-modified cells for adoptive cell therapy of cancer. Expert Opin. Biol. Ther. 2014, 14, 947–954. [Google Scholar] [CrossRef] [PubMed]
- 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. 2009, 33, 1255–1259. [Google Scholar] [CrossRef] [Green Version]
- Genßler, S.; Burger, M.C.; Zhang, C.; Oelsner, S.; Mildenberger, I.; Wagner, M.; Steinbach, J.P.; Wels, W.S. Dual targeting of glioblastoma with chimeric antigen receptor-engineered natural killer cells overcomes heterogeneity of target antigen expression and enhances antitumor activity and survival. Oncoimmunology 2015, 5, e1119354. [Google Scholar] [CrossRef] [PubMed]
- Liu, E.; Tong, Y.; Dotti, G.; Shaim, H.; Savoldo, B.; Mukherjee, M.; Orange, J.; Wan, X.; Lu, X.; Reynolds, A.; 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. [Google Scholar] [CrossRef] [PubMed]
- Marin, V.; Kakuda, H.; Dander, E.; Imai, C.; Campana, D.; Biondi, A.; D’Amico, G. Enhancement of the anti-leukemic activity of cytokine induced killer cells with an anti-CD19 chimeric receptor delivering a 4-1BB-zeta activating signal. Exp. Hematol. 2007, 35, 1388–1397. [Google Scholar] [CrossRef]
- Xu, Y.; Liu, Q.; Zhong, M.; Wang, Z.; Chen, Z.; Zhang, Y.; Xing, H.; Tian, Z.; Tang, K.; Liao, X.; et al. 2B4 costimulatory domain enhancing cytotoxic ability of anti-CD5 chimeric antigen receptor engineered natural killer cells against T cell malignancies. J. Hematol. Oncol. 2019, 12, 49. [Google Scholar] [CrossRef] [PubMed]
- Huang, Y.; Zeng, J.; Liu, T.; Xu, Q.; Song, X.; Zeng, J. DNAM1 and 2B4 Costimulatory Domains Enhance the Cytotoxicity of Anti-GPC3 Chimeric Antigen Receptor-Modified Natural Killer Cells Against Hepatocellular Cancer Cells in vitro. Cancer Manag. Res. 2020, 12, 3247–3255. [Google Scholar] [CrossRef]
- Marofi, F.; Rahman, H.S.; Thangavelu, L.; Dorofeev, A.; Bayas-Morejón, F.; Shirafkan, N.; Shomali, N.; Chartrand, M.S.; Jarahian, M.; Vahedi, G.; et al. Renaissance of armored immune effector cells, CAR-NK cells, brings the higher hope for successful cancer therapy. Stem Cell Res. Ther. 2021, 12, 200. [Google Scholar] [CrossRef] [PubMed]
- Imai, C.; Iwamoto, S.; Campana, D. Genetic modification of primary natural killer cells overcomes inhibitory signals and induces specific killing of leukemic cells. Blood 2005, 106, 376–383. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Töpfer, K.; Cartellieri, M.; Michen, S.; Wiedemuth, R.; Müller, N.; Lindemann, D.; Bachmann, M.; Füssel, M.; Schackert, G.; Temme, A. DAP12-based activating chimeric antigen receptor for NK cell tumor immunotherapy. J. Immunol. 2015, 194, 3201–3212. [Google Scholar] [CrossRef]
- Lanier, L.L. NK cell recognition. Annu. Rev. Immunol. 2005, 23, 225–274. [Google Scholar] [CrossRef]
- Xiao, L.; Cen, D.; Gan, H.; Sun, Y.; Huang, N.; Xiong, H.; Jin, Q.; Su, L.; Liu, X.; Wang, K.; et al. Adoptive Transfer of NKG2D CAR mRNA-Engineered Natural Killer Cells in Colorectal Cancer Patients. Mol. Ther. 2019, 27, 1114–1125. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Santomasso, B.; Bachier, C.; Westin, J.; Rezvani, K.; Shpall, E.J. The Other Side of CAR T-Cell Therapy: Cytokine Release Syndrome, Neurologic Toxicity, and Financial Burden. Am. Soc. Clin. Oncol. Educ. Book 2019, 39, 433–444. [Google Scholar] [CrossRef]
- Chou, C.K.; Turtle, C.J. Insight into mechanisms associated with cytokine release syndrome and neurotoxicity after CD19 CAR-T cell immunotherapy. Bone Marrow Transplant. 2019, 54, 780–784. [Google Scholar] [CrossRef]
- Klingemann, H. Are natural killer cells superior CAR drivers? Oncoimmunology 2014, 3, e28147. [Google Scholar] [CrossRef] [Green Version]
- Gill, S.; Olson, J.A.; Negrin, R.S. Natural killer cells in allogeneic transplantation: Effect on engraftment, graft-versus-tumor, and graft-versus-host responses. Biol. Blood Marrow Transplant. 2009, 15, 765–776. [Google Scholar] [CrossRef] [Green Version]
- Herrera, L.; Santos, S.; Vesga, M.A.; Anguita, J.; Martin-Ruiz, I.; Carrascosa, T.; Juan, M.; Eguizabal, C. Adult peripheral blood and umbilical cord blood NK cells are good sources for effective CAR therapy against CD19 positive leukemic cells. Sci. Rep. 2019, 9, 18729. [Google Scholar] [CrossRef] [Green Version]
- Olson, J.A.; Leveson-Gower, D.B.; Gill, S.; Baker, J.; Beilhack, A.; Negrin, R.S. NK cells mediate reduction of GVHD by inhibiting activated, alloreactive T cells while retaining GVT effects. Blood 2010, 115, 4293–4301. [Google Scholar] [CrossRef] [Green Version]
- Zhang, Y.; Wallace, D.L.; de Lara, C.M.; Ghattas, H.; Asquith, B.; Worth, A.; Griffin, G.E.; Taylor, G.P.; Tough, D.F.; Beverley, P.C.L.; et al. In vivo kinetics of human natural killer cells: The effects of ageing and acute and chronic viral infection. Immunology 2007, 121, 258–265. [Google Scholar] [CrossRef]
- Oei, V.Y.S.; Siernicka, M.; Graczyk-Jarzynka, A.; Hoel, H.J.; Yang, W.; Palacios, D.; Almåsbak, H.; Bajor, M.; Clement, D.; Brandt, L.; et al. Intrinsic Functional Potential of NK-Cell Subsets Constrains Retargeting Driven by Chimeric Antigen Receptors. Cancer Immunol. Res. 2018, 6, 467–480. [Google Scholar] [CrossRef] [Green Version]
- Majzner, R.G.; Mackall, C.L. Tumor Antigen Escape from CAR T-cell Therapy. Cancer Discov. 2018, 8, 1219–1226. [Google Scholar] [CrossRef] [Green Version]
- O’Rourke, D.M.; Nasrallah, M.P.; Desai, A.; Melenhorst, J.J.; Mansfield, K.; Morrissette, J.J.D.; Martinez-Lage, M.; Brem, S.; Maloney, E.; Shen, A.; et al. A single dose of peripherally infused EGFRvIII-directed CAR T cells mediates antigen loss and induces adaptive resistance in patients with recurrent glioblastoma. Sci. Transl. Med. 2017, 9, eaaa0984. [Google Scholar] [CrossRef] [Green Version]
- Burger, M.C.; Zhang, C.; Harter, P.N.; Romanski, A.; Strassheimer, F.; Senft, C.; Tonn, T.; Steinbach, J.P.; Wels, W.S. CAR-Engineered NK Cells for the Treatment of Glioblastoma: Turning Innate Effectors Into Precision Tools for Cancer Immunotherapy. Front. Immunol. 2019, 10, 2683. [Google Scholar] [CrossRef] [Green Version]
- Jiang, H.; Zhang, W.; Shang, P.; Zhang, H.; Fu, W.; Ye, F.; Zeng, T.; Huang, H.; Zhang, X.; Sun, W.; et al. Transfection of chimeric anti-CD138 gene enhances natural killer cell activation and killing of multiple myeloma cells. Mol. Oncol. 2014, 8, 297–310. [Google Scholar] [CrossRef]
- Zhang, C.; Burger, M.C.; Jennewein, L.; Genßler, S.; Schönfeld, K.; Zeiner, P.; Hattingen, E.; Harter, P.N.; Mittelbronn, M.; Tonn, T.; et al. ErbB2/HER2-Specific NK Cells for Targeted Therapy of Glioblastoma. J. Natl. Cancer Inst. 2016, 108. [Google Scholar] [CrossRef]
- Cao, B.; Liu, M.; Wang, L.; Liang, B.; Feng, Y.; Chen, X.; Shi, Y.; Zhang, J.; Ye, X.; Tian, Y.; et al. Use of chimeric antigen receptor NK-92 cells to target mesothelin in ovarian cancer. Biochem. Biophys. Res. Commun. 2020, 524, 96–102. [Google Scholar] [CrossRef]
- Zhang, Q.; Tian, K.; Xu, J.; Zhang, H.; Li, L.; Fu, Q.; Chai, D.; Li, H.; Zheng, J. Synergistic Effects of Cabozantinib and EGFR-Specific CAR-NK-92 Cells in Renal Cell Carcinoma. J. Immunol. Res. 2017, 2017, 6915912. [Google Scholar] [CrossRef]
- Han, J.; Chu, J.; Keung Chan, W.; Zhang, J.; Wang, Y.; Cohen, J.B.; Victor, A.; Meisen, W.H.; Kim, S.-H.; Grandi, P.; et al. CAR-Engineered NK Cells Targeting Wild-Type EGFR and EGFRvIII Enhance Killing of Glioblastoma and Patient-Derived Glioblastoma Stem Cells. Sci. Rep. 2015, 5, 11483. [Google Scholar] [CrossRef]
- Murakami, T.; Nakazawa, T.; Natsume, A.; Nishimura, F.; Nakamura, M.; Matsuda, R.; Omoto, K.; Tanaka, Y.; Shida, Y.; Park, Y.-S.; et al. Novel Human NK Cell Line Carrying CAR Targeting EGFRvIII Induces Antitumor Effects in Glioblastoma Cells. Anticancer Res. 2018, 38, 5049–5056. [Google Scholar] [CrossRef]
- Chen, X.; Han, J.; Chu, J.; Zhang, L.; Zhang, J.; Chen, C.; Chen, L.; Wang, Y.; Wang, H.; Yi, L.; et al. A combinational therapy of EGFR-CAR NK cells and oncolytic herpes simplex virus 1 for breast cancer brain metastases. Oncotarget 2016, 7, 27764–27777. [Google Scholar] [CrossRef] [Green Version]
- Xia, N.; Haopeng, P.; Gong, J.U.; Lu, J.; Chen, Z.; Zheng, Y.; Wang, Z.; Sun, Y.U.; Yang, Z.; Hoffman, R.M.; et al. Robo1-specific CAR-NK Immunotherapy Enhances Efficacy of (125)I Seed Brachytherapy in an Orthotopic Mouse Model of Human Pancreatic Carcinoma. Anticancer Res. 2019, 39, 5919–5925. [Google Scholar] [CrossRef] [Green Version]
- Qu, Y.; Bi, J.Z. Killing effect of Robo1 targeted Chimeric Antigen Receptor modified NK92 cells against glioma and neuroblastoma cells. Zhonghua Yi Xue Za Zhi 2018, 98, 860–866. [Google Scholar] [CrossRef]
- Bigner, S.H.; Humphrey, P.A.; Wong, A.J.; Vogelstein, B.; Mark, J.; Friedman, H.S.; Bigner, D.D. Characterization of the epidermal growth factor receptor in human glioma cell lines and xenografts. Cancer Res. 1990, 50, 8017–8022. [Google Scholar]
- Aldape, K.D.; Ballman, K.; Furth, A.; Buckner, J.C.; Giannini, C.; Burger, P.C.; Scheithauer, B.W.; Jenkins, R.B.; James, C.D. Immunohistochemical detection of EGFRvIII in high malignancy grade astrocytomas and evaluation of prognostic significance. J. Neuropathol. Exp. Neurol. 2004, 63, 700–707. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Brennan, C.W.; Verhaak, R.G.W.; McKenna, A.; Campos, B.; Noushmehr, H.; Salama, S.R.; Zheng, S.; Chakravarty, D.; Sanborn, J.Z.; Berman, S.H.; et al. The somatic genomic landscape of glioblastoma. Cell 2013, 155, 462–477. [Google Scholar] [CrossRef]
- Nakazawa, T.; Murakami, T.; Natsume, A.; Nishimura, F.; Morimoto, T.; Matsuda, R.; Nakamura, M.; Yamada, S.; Nakagawa, I.; Park, Y.-S.; et al. KHYG-1 Cells with EGFRvIII-specific CAR Induced a Pseudoprogression-like Feature in Subcutaneous Tumours Derived from Glioblastoma-like Cells. Anticancer Res. 2020, 40, 3231–3237. [Google Scholar] [CrossRef]
- Müller, N.; Michen, S.; Tietze, S.; Töpfer, K.; Schulte, A.; Lamszus, K.; Schmitz, M.; Schackert, G.; Pastan, I.; Temme, A. Engineering NK Cells Modified with an EGFRvIII-specific Chimeric Antigen Receptor to Overexpress CXCR4 Improves Immunotherapy of CXCL12/SDF-1α-secreting Glioblastoma. J. Immunother. 2015, 38, 197–210. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Ahmed, N.; Salsman, V.S.; Kew, Y.; Shaffer, D.; Powell, S.; Zhang, Y.J.; Grossman, R.G.; Heslop, H.E.; Gottschalk, S. HER2-specific T cells target primary glioblastoma stem cells and induce regression of autologous experimental tumors. Clin. Cancer Res. 2010, 16, 474–485. [Google Scholar] [CrossRef] [Green Version]
- Koka, V.; Potti, A.; Forseen, S.E.; Pervez, H.; Fraiman, G.N.; Koch, M.; Levitt, R. Role of Her-2/neu overexpression and clinical determinants of early mortality in glioblastoma multiforme. Am. J. Clin. Oncol. 2003, 26, 332–335. [Google Scholar] [CrossRef]
- Schlegel, J.; Stumm, G.; Brändle, K.; Merdes, A.; Mechtersheimer, G.; Hynes, N.E.; Kiessling, M. Amplification and differential expression of members of the erbB-gene family in human glioblastoma. J. Neurooncol. 1994, 22, 201–207. [Google Scholar] [CrossRef]
- Burger, M.C.; Mildenberger, I.; Zhang, C.; Ihrig, K.; Wagner, M.; Mittelbronn, M.; Senft, C.; Tonn, T.; Wels, W.S.; Steinbach, J.P. P04.05 The CAR2BRAIN study: A monocentric phase I trial with ErbB2-specific NK-92/5.28.z cells in recurrent glioblastoma. Neuro-Oncol. 2016, 18, iv24–iv25. [Google Scholar] [CrossRef] [Green Version]
- Carlsten, M.; Childs, R.W. Genetic Manipulation of NK Cells for Cancer Immunotherapy: Techniques and Clinical Implications. Front. Immunol. 2015, 6, 266. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Colamartino, A.B.L.; Lemieux, W.; Bifsha, P.; Nicoletti, S.; Chakravarti, N.; Sanz, J.; Roméro, H.; Selleri, S.; Béland, K.; Guiot, M.; et al. Efficient and Robust NK-Cell Transduction with Baboon Envelope Pseudotyped Lentivector. Front. Immunol. 2019, 10, 2873. [Google Scholar] [CrossRef] [PubMed]
- Fenard, D.; Ingrao, D.; Seye, A.; Buisset, J.; Genries, S.; Martin, S.; Kichler, A.; Galy, A. Vectofusin-1, a new viral entry enhancer, strongly promotes lentiviral transduction of human hematopoietic stem cells. Mol. Ther. Nucleic Acids 2013, 2, e90. [Google Scholar] [CrossRef]
- Papayannakos, C.; Daniel, R. Understanding lentiviral vector chromatin targeting: Working to reduce insertional mutagenic potential for gene therapy. Gene Ther. 2013, 20, 581–588. [Google Scholar] [CrossRef] [Green Version]
- Li, Y.; Hermanson, D.L.; Moriarity, B.S.; Kaufman, D.S. Human iPSC-Derived Natural Killer Cells Engineered with Chimeric Antigen Receptors Enhance Anti-tumor Activity. Cell Stem Cell 2018, 23, 181–192.e5. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Huang, R.-S.; Shih, H.-A.; Lai, M.-C.; Chang, Y.-J.; Lin, S. Enhanced NK-92 Cytotoxicity by CRISPR Genome Engineering Using Cas9 Ribonucleoproteins. Front. Immunol. 2020, 11, 1008. [Google Scholar] [CrossRef]
- Meazza, R.; Azzarone, B.; Orengo, A.M.; Ferrini, S. Role of common-gamma chain cytokines in NK cell development and function: Perspectives for immunotherapy. J. Biomed. Biotechnol. 2011, 2011, 861920. [Google Scholar] [CrossRef] [PubMed]
- Xu, X.; Huang, W.; Heczey, A.; Liu, D.; Guo, L.; Wood, M.; Jin, J.; Courtney, A.N.; Liu, B.; Di Pierro, E.J.; et al. NKT Cells Coexpressing a GD2-Specific Chimeric Antigen Receptor and IL15 Show Enhanced In Vivo Persistence and Antitumor Activity against Neuroblastoma. Clin. Cancer Res. 2019, 25, 7126–7138. [Google Scholar] [CrossRef] [Green Version]
- Yvon, E.S.; Burga, R.; Powell, A.; Cruz, C.R.; Fernandes, R.; Barese, C.; Nguyen, T.; Abdel-Baki, M.S.; Bollard, C.M. Cord blood natural killer cells expressing a dominant negative TGF-β receptor: Implications for adoptive immunotherapy for glioblastoma. Cytotherapy 2017, 19, 408–418. [Google Scholar] [CrossRef] [Green Version]
- Yang, B.; Liu, H.; Shi, W.; Wang, Z.; Sun, S.; Zhang, G.; Hu, Y.; Liu, T.; Jiao, S. Blocking transforming growth factor-β signaling pathway augments antitumor effect of adoptive NK-92 cell therapy. Int. Immunopharmacol. 2013, 17, 198–204. [Google Scholar] [CrossRef]
- Rosenberg, S.A.; Lotze, M.T.; Muul, L.M.; Leitman, S.; Chang, A.E.; Ettinghausen, S.E.; Matory, Y.L.; Skibber, J.M.; Shiloni, E.; Vetto, J.T. Observations on the systemic administration of autologous lymphokine-activated killer cells and recombinant interleukin-2 to patients with metastatic cancer. N. Engl. J. Med. 1985, 313, 1485–1492. [Google Scholar] [CrossRef]
- Phillips, J.H.; Lanier, L.L. Dissection of the lymphokine-activated killer phenomenon. Relative contribution of peripheral blood natural killer cells and T lymphocytes to cytolysis. J. Exp. Med. 1986, 164, 814–825. [Google Scholar] [CrossRef]
- Gaffen, S.L.; Liu, K.D. Overview of interleukin-2 function, production and clinical applications. Cytokine 2004, 28, 109–123. [Google Scholar] [CrossRef] [PubMed]
- West, W.H.; Tauer, K.W.; Yannelli, J.R.; Marshall, G.D.; Orr, D.W.; Thurman, G.B.; Oldham, R.K. Constant-infusion recombinant interleukin-2 in adoptive immunotherapy of advanced cancer. N. Engl. J. Med. 1987, 316, 898–905. [Google Scholar] [CrossRef] [PubMed]
- Schwartzentruber, D.J. High-dose interleukin-2 is an intensive treatment regardless of the venue of administration. Cancer J. 2001, 7, 103–104. [Google Scholar]
- 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. 2015, 42, 539–548. [Google Scholar] [CrossRef] [Green Version]
- Childs, R.W.; Carlsten, M. Therapeutic approaches to enhance natural killer cell cytotoxicity against cancer: The force awakens. Nat. Rev. Drug Discov. 2015, 14, 487–498. [Google Scholar] [CrossRef] [PubMed]
- Levin, A.M.; Bates, D.L.; Ring, A.M.; Krieg, C.; Lin, J.T.; Su, L.; Moraga, I.; Raeber, M.E.; Bowman, G.R.; Novick, P.; et al. Exploiting a natural conformational switch to engineer an interleukin-2 “superkine”. Nature 2012, 484, 529–533. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Mitra, S.; Ring, A.M.; Amarnath, S.; Spangler, J.B.; Li, P.; Ju, W.; Fischer, S.; Oh, J.; Spolski, R.; Weiskopf, K.; et al. Interleukin-2 activity can be fine tuned with engineered receptor signaling clamps. Immunity 2015, 42, 826–838. [Google Scholar] [CrossRef] [Green Version]
- Sakamoto, N.; Ishikawa, T.; Kokura, S.; Okayama, T.; Oka, K.; Ideno, M.; Sakai, F.; Kato, A.; Tanabe, M.; Enoki, T.; 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. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Szmania, S.; Lapteva, N.; Garg, T.; Greenway, A.; Lingo, J.; Nair, B.; Stone, K.; Woods, E.; Khan, J.; Stivers, J.; et al. Ex vivo-expanded natural killer cells demonstrate robust proliferation in vivo in high-risk relapsed multiple myeloma patients. J. Immunother. 2015, 38, 24–36. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Shaffer, B.C.; Le Luduec, J.-B.; Forlenza, C.; Jakubowski, A.A.; Perales, M.-A.; Young, J.W.; Hsu, K.C. Phase II Study of Haploidentical Natural Killer Cell Infusion for Treatment of Relapsed or Persistent Myeloid Malignancies Following Allogeneic Hematopoietic Cell Transplantation. Biol. Blood Marrow Transplant. 2016, 22, 705–709. [Google Scholar] [CrossRef] [Green Version]
- Colombo, F.; Barzon, L.; Franchin, E.; Pacenti, M.; Pinna, V.; Danieli, D.; Zanusso, M.; Palù, G. Combined HSV-TK/IL-2 gene therapy in patients with recurrent glioblastoma multiforme: Biological and clinical results. Cancer Gene Ther. 2005, 12, 835–848. [Google Scholar] [CrossRef] [PubMed]
- Qiao, H.B.; Li, J.; Lv, L.-J.; Nie, B.-J.; Lu, P.; Xue, F.; Zhang, Z.-M. The effects of interleukin 2 and rAd-p53 as a treatment for glioblastoma. Mol. Med. Rep. 2018, 17, 4853–4859. [Google Scholar] [CrossRef] [PubMed]
- Conlon, K.C.; Lugli, E.; Welles, H.C.; Rosenberg, S.A.; Fojo, A.T.; Morris, J.C.; Fleisher, T.A.; Dubois, S.P.; Perera, L.P.; Stewart, D.M.; et al. Redistribution, hyperproliferation, activation of natural killer cells and CD8 T cells, and cytokine production during first-in-human clinical trial of recombinant human interleukin-15 in patients with cancer. J. Clin. Oncol. 2015, 33, 74–82. [Google Scholar] [CrossRef] [PubMed]
- Miller, J.S.; Morishima, C.; McNeel, D.G.; Patel, M.R.; Kohrt, H.E.K.; Thompson, J.A.; Sondel, P.M.; Wakelee, H.A.; Disis, M.L.; Kaiser, J.C.; et al. A First-in-Human Phase I Study of Subcutaneous Outpatient Recombinant Human IL15 (rhIL15) in Adults with Advanced Solid Tumors. Clin. Cancer Res. 2018, 24, 1525–1535. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Prlic, M.; Blazar, B.R.; Farrar, M.A.; Jameson, S.C. In vivo survival and homeostatic proliferation of natural killer cells. J. Exp. Med. 2003, 197, 967–976. [Google Scholar] [CrossRef]
- Fehniger, T.A.; Caligiuri, M.A. Interleukin 15: Biology and relevance to human disease. Blood 2001, 97, 14–32. [Google Scholar] [CrossRef]
- Leclercq, G.; Debacker, V.; de Smedt, M.; Plum, J. Differential effects of interleukin-15 and interleukin-2 on differentiation of bipotential T/natural killer progenitor cells. J. Exp. Med. 1996, 184, 325–336. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Pillet, A.-H.; Bugault, F.; Thèze, J.; Chakrabarti, L.A.; Rose, T. A programmed switch from IL-15- to IL-2-dependent activation in human NK cells. J. Immunol. 2009, 182, 6267–6277. [Google Scholar] [CrossRef] [PubMed]
- Pillet, A.-H.; Thèze, J.; Rose, T. Interleukin (IL)-2 and IL-15 have different effects on human natural killer lymphocytes. Hum. Immunol. 2011, 72, 1013–1017. [Google Scholar] [CrossRef] [PubMed]
- Stoklasek, T.A.; Schluns, K.S.; Lefrançois, L. Combined IL-15/IL-15Ralpha immunotherapy maximizes IL-15 activity in vivo. J. Immunol. 2006, 177, 6072–6080. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Xu, W.; Jones, M.; Liu, B.; Zhu, X.; Johnson, C.B.; Edwards, A.C.; Kong, L.; Jeng, E.K.; Han, K.; Marcus, W.D.; et al. Efficacy and mechanism-of-action of a novel superagonist interleukin-15: Interleukin-15 receptor αSu/Fc fusion complex in syngeneic murine models of multiple myeloma. Cancer Res. 2013, 73, 3075–3086. [Google Scholar] [CrossRef] [Green Version]
- Dubois, S.; Patel, H.J.; Zhang, M.; Waldmann, T.A.; Müller, J.R. Preassociation of IL-15 with IL-15R alpha-IgG1-Fc enhances its activity on proliferation of NK and CD8+/CD44high T cells and its antitumor action. J. Immunol. 2008, 180, 2099–2106. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Chen, Y.; Chen, B.; Yang, T.; Xiao, W.; Qian, L.; Ding, Y.; Ji, M.; Ge, X.; Gong, W. Human fused NKG2D-IL-15 protein controls xenografted human gastric cancer through the recruitment and activation of NK cells. Cell. Mol. Immunol. 2017, 14, 293–307. [Google Scholar] [CrossRef] [PubMed]
- Thaysen-Andersen, M.; Chertova, E.; Bergamaschi, C.; Moh, E.S.X.; Chertov, O.; Roser, J.; Sowder, R.; Bear, J.; Lifson, J.; Packer, N.H.; et al. Recombinant human heterodimeric IL-15 complex displays extensive and reproducible N- and O-linked glycosylation. Glycoconj. J. 2016, 33, 417–433. [Google Scholar] [CrossRef]
- Rosario, M.; Liu, B.; Kong, L.; Collins, L.I.; Schneider, S.E.; Chen, X.; Han, K.; Jeng, E.K.; Rhode, P.R.; Leong, J.W.; et al. The IL-15-Based ALT-803 Complex Enhances FcγRIIIa-Triggered NK Cell Responses and In Vivo Clearance of B Cell Lymphomas. Clin. Cancer Res. 2016, 22, 596–608. [Google Scholar] [CrossRef] [Green Version]
- Han, K.; Zhu, X.; Liu, B.; Jeng, E.; Kong, L.; Yovandich, J.L.; Vyas, V.V.; Marcus, W.D.; Chavaillaz, P.-A.; Romero, C.A.; et al. IL-15: IL-15 receptor alpha superagonist complex: High-level co-expression in recombinant mammalian cells, purification and characterization. Cytokine 2011, 56, 804–810. [Google Scholar] [CrossRef] [Green Version]
- Margolin, K.; Morishima, C.; Velcheti, V.; Miller, J.S.; Lee, S.M.; Silk, A.W.; Holtan, S.G.; Lacroix, A.M.; Fling, S.P.; Kaiser, J.C.; et al. Phase I Trial of ALT-803, A Novel Recombinant IL15 Complex, in Patients with Advanced Solid Tumors. Clin. Cancer Res. 2018, 24, 5552–5561. [Google Scholar] [CrossRef] [Green Version]
- Conlon, K.C.; Potter, E.L.; Pittaluga, S.; Lee, C.-C.R.; Miljkovic, M.D.; Fleisher, T.A.; Dubois, S.; Bryant, B.R.; Petrus, M.; Perera, L.P.; et al. IL15 by Continuous Intravenous Infusion to Adult Patients with Solid Tumors in a Phase I Trial Induced Dramatic NK-Cell Subset Expansion. Clin. Cancer Res. 2019, 25, 4945–4954. [Google Scholar] [CrossRef] [PubMed]
- Dubois, S.P.; Miljkovic, M.D.; Fleisher, T.A.; Pittaluga, S.; Hsu-Albert, J.; Bryant, B.R.; Petrus, M.N.; Perera, L.P.; Müller, J.R.; Shih, J.H.; et al. Short-course IL-15 given as a continuous infusion led to a massive expansion of effective NK cells: Implications for combination therapy with antitumor antibodies. J. Immunother. Cancer 2021, 9, e002193. [Google Scholar] [CrossRef]
- Schmohl, J.U.; Felices, M.; Taras, E.; Miller, J.S.; Vallera, D.A. Enhanced ADCC and NK Cell Activation of an Anticarcinoma Bispecific Antibody by Genetic Insertion of a Modified IL-15 Cross-linker. Mol. Ther. 2016, 24, 1312–1322. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Krenciute, G.; Prinzing, B.L.; Yi, Z.; Wu, M.-F.; Liu, H.; Dotti, G.; Balyasnikova, I.V.; Gottschalk, S. Transgenic Expression of IL15 Improves Antiglioma Activity of IL13Rα2-CAR T Cells but Results in Antigen Loss Variants. Cancer Immunol. Res. 2017, 5, 571–581. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Zhang, Q.; Zhang, J.; Tian, Y.; Zhu, G.; Liu, S.; Liu, F. Efficacy of a novel double-controlled oncolytic adenovirus driven by the Ki67 core promoter and armed with IL-15 against glioblastoma cells. Cell Biosci. 2020, 10, 124. [Google Scholar] [CrossRef]
- Yiang, G.-T.; Chou, R.-H.; Chang, W.-J.; Wei, C.-W.; Yu, Y.-L. Long-term expression of rAAV2-hIL15 enhances immunoglobulin production and lymphokine-activated killer cell-mediated human glioblastoma cell death. Mol. Clin. Oncol. 2013, 1, 321–325. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Ma, R.; Lu, T.; Li, Z.; Teng, K.-Y.; Mansour, A.G.; Yu, M.; Tian, L.; Xu, B.; Ma, S.; Zhang, J.; et al. An oncolytic virus expressing IL-15/IL-15Rα combined with off-the-shelf EGFR-CAR NK cells targets glioblastoma. Cancer Res. 2021, 81, 3635–3648. [Google Scholar] [CrossRef] [PubMed]
- Mathios, D.; Park, C.-K.; Marcus, W.D.; Alter, S.; Rhode, P.R.; Jeng, E.K.; Wong, H.C.; Pardoll, D.M.; Lim, M. 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. [Google Scholar] [CrossRef]
- Gomes-Giacoia, E.; Miyake, M.; Goodison, S.; Sriharan, A.; Zhang, G.; You, L.; Egan, J.O.; Rhode, P.R.; Parker, A.S.; Chai, K.X.; et al. Intravesical ALT-803 and BCG treatment reduces tumor burden in a carcinogen induced bladder cancer rat model; a role for cytokine production and NK cell expansion. PLoS ONE 2014, 9, e96705. [Google Scholar] [CrossRef]
- Zhu, L.; Kalimuthu, S.; Oh, J.M.; Gangadaran, P.; Baek, S.H.; Jeong, S.Y.; Lee, S.-W.; Lee, J.; Ahn, B.-C. Enhancement of antitumor potency of extracellular vesicles derived from natural killer cells by IL-15 priming. Biomaterials 2019, 190–191, 38–50. [Google Scholar] [CrossRef]
- Smyth, M.J.; Taniguchi, M.; Street, S.E. The anti-tumor activity of IL-12: Mechanisms of innate immunity that are model and dose dependent. J. Immunol. 2000, 165, 2665–2670. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Sahin, U.; Kraft-Bauer, S.; Ohnesorge, S.; Pfreundschuh, M.; Renner, C. Interleukin-12 increases bispecific-antibody-mediated natural killer cell cytotoxicity against human tumors. Cancer Immunol. Immunother. 1996, 42, 9–14. [Google Scholar] [CrossRef]
- Parihar, R.; Nadella, P.; Lewis, A.; Jensen, R.; De Hoff, C.; Dierksheide, J.E.; VanBuskirk, A.M.; Magro, C.M.; Young, D.C.; Shapiro, C.L.; et al. 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. 2004, 10, 5027–5037. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Gokhale, M.S.; Vainstein, V.; Tom, J.; Thomas, S.; Lawrence, C.E.; Gluzman-Poltorak, Z.; Siebers, N.; Basile, L.A. Single low-dose rHuIL-12 safely triggers multilineage hematopoietic and immune-mediated effects. Exp. Hematol. Oncol. 2014, 3, 11. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Basile, L.A.; Gallaher, T.K.; Shibata, D.; Miller, J.D.; Douer, D. Multilineage hematopoietic recovery with concomitant antitumor effects using low dose Interleukin-12 in myelosuppressed tumor-bearing mice. J. Transl. Med. 2008, 6, 26. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Fallon, J.; Tighe, R.; Kradjian, G.; Guzman, W.; Bernhardt, A.; Neuteboom, B.; Lan, Y.; Sabzevari, H.; Schlom, J.; Greiner, J.W. The immunocytokine NHS-IL12 as a potential cancer therapeutic. Oncotarget 2014, 5, 1869–1884. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Agliardi, G.; Liuzzi, A.R.; Hotblack, A.; De Feo, D.; Núñez, N.; Stowe, C.L.; Friebel, E.; Nannini, F.; Rindlisbacher, L.; Roberts, T.A.; et al. Intratumoral IL-12 delivery empowers CAR-T cell immunotherapy in a pre-clinical model of glioblastoma. Nat. Commun. 2021, 12, 444. [Google Scholar] [CrossRef] [PubMed]
- Brempelis, K.J.; Cowan, C.M.; Kreuser, S.A.; Labadie, K.P.; Prieskorn, B.M.; Lieberman, N.A.P.; Ene, C.I.; Moyes, K.W.; Chinn, H.; DeGolier, K.R.; et al. Genetically engineered macrophages persist in solid tumors and locally deliver therapeutic proteins to activate immune responses. J. Immunother. Cancer 2020, 8, e001356. [Google Scholar] [CrossRef]
- Chiocca, E.A.; Yu, J.S.; Lukas, R.V.; Solomon, I.H.; Ligon, K.L.; Nakashima, H.; Triggs, D.A.; Reardon, D.A.; Wen, P.; Stopa, B.M.; et al. Regulatable interleukin-12 gene therapy in patients with recurrent high-grade glioma: Results of a phase 1 trial. Sci. Transl. Med. 2019, 11, eaaw5680. [Google Scholar] [CrossRef] [PubMed]
- Patel, D.M.; Foreman, P.M.; Nabors, L.B.; Riley, K.O.; Gillespie, G.Y.; Markert, J.M. Design of a Phase I Clinical Trial to Evaluate M032, a Genetically Engineered HSV-1 Expressing IL-12, in Patients with Recurrent/Progressive Glioblastoma Multiforme, Anaplastic Astrocytoma, or Gliosarcoma. Hum. Gene Ther. Clin. Dev. 2016, 27, 69–78. [Google Scholar] [CrossRef]
- Chiu, T.-L.; Peng, C.-W.; Wang, M.-J. Enhanced anti-glioblastoma activity of microglia by AAV2-mediated IL-12 through TRAIL and phagocytosis in vitro. Oncol. Rep. 2011, 25, 1373–1380. [Google Scholar] [CrossRef]
- Chiu, T.-L.; Lin, S.-Z.; Hsieh, W.-H.; Peng, C.-W. AAV2-mediated interleukin-12 in the treatment of malignant brain tumors through activation of NK cells. Int. J. Oncol. 2009, 35, 1361–1367. [Google Scholar] [CrossRef] [Green Version]
- Zhang, W.; Fulci, G.; Wakimoto, H.; Cheema, T.A.; Buhrman, J.S.; Jeyaretna, D.S.; Stemmer Rachamimov, A.O.; Rabkin, S.D.; Martuza, R.L. Combination of oncolytic herpes simplex viruses armed with angiostatin and IL-12 enhances antitumor efficacy in human glioblastoma models. Neoplasia 2013, 15, 591–599. [Google Scholar] [CrossRef] [Green Version]
- Vom Berg, J.; Vrohlings, M.; Haller, S.; Haimovici, A.; Kulig, P.; Sledzinska, A.; Weller, M.; Becher, B. Intratumoral IL-12 combined with CTLA-4 blockade elicits T cell-mediated glioma rejection. J. Exp. Med. 2013, 210, 2803–2811. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Kasaian, M.T.; Whitters, M.J.; Carter, L.L.; Lowe, L.D.; Jussif, J.M.; Deng, B.; Johnson, K.A.; Witek, J.S.; Senices, M.; Konz, R.F.; et al. IL-21 limits NK cell responses and promotes antigen-specific T cell activation: A mediator of the transition from innate to adaptive immunity. Immunity 2002, 16, 559–569. [Google Scholar] [CrossRef] [Green Version]
- Ma, H.-L.; Whitters, M.J.; Konz, R.F.; Senices, M.; Young, D.A.; Grusby, M.J.; Collins, M.; Dunussi-Joannopoulos, K. IL-21 activates both innate and adaptive immunity to generate potent antitumor responses that require perforin but are independent of IFN-gamma. J. Immunol. 2003, 171, 608–615. [Google Scholar] [CrossRef] [Green Version]
- He, H.; Wisner, P.; Yang, G.; Hu, H.-M.; Haley, D.; Miller, W.; O’Hara, A.; Alvord, W.G.; Clegg, C.H.; Fox, B.A.; et al. Combined IL-21 and low-dose IL-2 therapy induces anti-tumor immunity and long-term curative effects in a murine melanoma tumor model. J. Transl. Med. 2006, 4, 24. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- McMichael, E.L.; Jaime-Ramirez, A.C.; Guenterberg, K.D.; Luedke, E.; Atwal, L.S.; Campbell, A.R.; Hu, Z.; Tatum, A.S.; Kondadasula, S.V.; Mo, X.; et al. IL-21 Enhances Natural Killer Cell Response to Cetuximab-Coated Pancreatic Tumor Cells. Clin. Cancer Res. 2017, 23, 489–502. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Petrella, T.M.; Mihalcioiu, C.L.D.; McWhirter, E.; Belanger, K.; Savage, K.J.; Song, X.; Hamid, O.; Cheng, T.; Davis, M.L.; Lee, C.W.; et al. Final efficacy results of NCIC CTG IND.202: A randomized phase II study of recombinant interleukin-21 (rIL21) in patients with recurrent or metastatic melanoma (MM). J. Clin. Oncol. 2013, 31, 9032. [Google Scholar] [CrossRef]
- Petrella, T.M.; Tozer, R.; Belanger, K.; Savage, K.J.; Wong, R.; Smylie, M.; Kamel-Reid, S.; Tron, V.; Chen, B.E.; Hunder, N.N.; et al. Interleukin-21 has activity in patients with metastatic melanoma: A phase II study. J. Clin. Oncol. 2012, 30, 3396–3401. [Google Scholar] [CrossRef] [PubMed]
- Bhatia, S.; Curti, B.; Ernstoff, M.S.; Gordon, M.; Heath, E.I.; Miller, W.H.J.; Puzanov, I.; Quinn, D.I.; Flaig, T.W.; VanVeldhuizen, P.; et al. Recombinant interleukin-21 plus sorafenib for metastatic renal cell carcinoma: A phase 1/2 study. J. Immunother. Cancer 2014, 2, 2. [Google Scholar] [CrossRef] [Green Version]
- Grünwald, V.; Desar, I.M.E.; Haanen, J.; Fiedler, W.; Mouritzen, U.; Olsen, M.W.B.; van Herpen, C.M.L. A phase I study of recombinant human interleukin-21 (rIL-21) in combination with sunitinib in patients with metastatic renal cell carcinoma (RCC). Acta Oncol. 2011, 50, 121–126. [Google Scholar] [CrossRef] [PubMed]
- Timmerman, J.M.; Byrd, J.C.; Andorsky, D.J.; Yamada, R.E.; Kramer, J.; Muthusamy, N.; Hunder, N.; Pagel, J.M. A phase I dose-finding trial of recombinant interleukin-21 and rituximab in relapsed and refractory low grade B-cell lymphoproliferative disorders. Clin. Cancer Res. 2012, 18, 5752–5760. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Steele, N.; Anthony, A.; Saunders, M.; Esmarck, B.; Ehrnrooth, E.; Kristjansen, P.E.G.; Nihlén, A.; Hansen, L.T.; Cassidy, J. A phase 1 trial of recombinant human IL-21 in combination with cetuximab in patients with metastatic colorectal cancer. Br. J. Cancer 2012, 106, 793–798. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Daga, A.; Orengo, A.M.; Gangemi, R.M.R.; Marubbi, D.; Perera, M.; Comes, A.; Ferrini, S.; Corte, G. Glioma immunotherapy by IL-21 gene-modified cells or by recombinant IL-21 involves antibody responses. Int. J. Cancer 2007, 121, 1756–1763. [Google Scholar] [CrossRef]
- Joalland, N.; Chauvin, C.; Oliver, L.; Vallette, F.M.; Pecqueur, C.; Jarry, U.; Scotet, E. IL-21 Increases the Reactivity of Allogeneic Human Vγ9Vδ2 T Cells Against Primary Glioblastoma Tumors. J. Immunother. 2018, 41, 224–231. [Google Scholar] [CrossRef]
- Wölfl, M.; Merker, K.; Morbach, H.; Van Gool, S.W.; Eyrich, M.; Greenberg, P.D.; Schlegel, P.G. Primed tumor-reactive multifunctional CD62L+ human CD8+ T cells for immunotherapy. Cancer Immunol. Immunother. 2011, 60, 173–186. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Fehniger, T.A.; Shah, M.H.; Turner, M.J.; VanDeusen, J.B.; Whitman, S.P.; Cooper, M.A.; Suzuki, K.; Wechser, M.; Goodsaid, F.; Caligiuri, M.A. Differential cytokine and chemokine gene expression by human NK cells following activation with IL-18 or IL-15 in combination with IL-12: Implications for the innate immune response. J. Immunol. 1999, 162, 4511–4520. [Google Scholar]
- Srivastava, S.; Pelloso, D.; Feng, H.; Voiles, L.; Lewis, D.; Haskova, Z.; Whitacre, M.; Trulli, S.; Chen, Y.-J.; Toso, J.; et al. Effects of interleukin-18 on natural killer cells: Costimulation of activation through Fc receptors for immunoglobulin. Cancer Immunol. Immunother. 2013, 62, 1073–1082. [Google Scholar] [CrossRef] [Green Version]
- Nielsen, C.M.; Wolf, A.-S.; Goodier, M.R.; Riley, E.M. Synergy between Common γ Chain Family Cytokines and IL-18 Potentiates Innate and Adaptive Pathways of NK Cell Activation. Front. Immunol. 2016, 7, 101. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- 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. 2012, 209, 2351–2365. [Google Scholar] [CrossRef]
- Scott, A.M.; Wolchok, J.D.; Old, L.J. Antibody therapy of cancer. Nat. Rev. Cancer 2012, 12, 278–287. [Google Scholar] [CrossRef] [PubMed]
- Patel, D.; Lahiji, A.; Patel, S.; Franklin, M.; Jimenez, X.; Hicklin, D.J.; Kang, X. Monoclonal antibody cetuximab binds to and down-regulates constitutively activated epidermal growth factor receptor vIII on the cell surface. Anticancer Res. 2007, 27, 3355–3366. [Google Scholar]
- Fukai, J.; Nishio, K.; Itakura, T.; Koizumi, F. Antitumor activity of cetuximab against malignant glioma cells overexpressing EGFR deletion mutant variant III. Cancer Sci. 2008, 99, 2062–2069. [Google Scholar] [CrossRef]
- Neyns, B.; Sadones, J.; Joosens, E.; Bouttens, F.; Verbeke, L.; Baurain, J.-F.; D’Hondt, L.; Strauven, T.; Chaskis, C.; In’t Veld, P.; et al. Stratified phase II trial of cetuximab in patients with recurrent high-grade glioma. Ann. Oncol. 2009, 20, 1596–1603. [Google Scholar] [CrossRef]
- Shields, R.L.; Namenuk, A.K.; Hong, K.; Meng, Y.G.; Rae, J.; Briggs, J.; Xie, D.; Lai, J.; Stadlen, A.; Li, B.; et al. High resolution mapping of the binding site on human IgG1 for Fc gamma RI, Fc gamma RII, Fc gamma RIII, and FcRn and design of IgG1 variants with improved binding to the Fc gamma R. J. Biol. Chem. 2001, 276, 6591–6604. [Google Scholar] [CrossRef] [Green Version]
- Lazar, G.A.; Dang, W.; Karki, S.; Vafa, O.; Peng, J.S.; Hyun, L.; Chan, C.; Chung, H.S.; Eivazi, A.; Yoder, S.C.; et al. Engineered antibody Fc variants with enhanced effector function. Proc. Natl. Acad. Sci. USA 2006, 103, 4005–4010. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Stavenhagen, J.B.; Gorlatov, S.; Tuaillon, N.; Rankin, C.T.; Li, H.; Burke, S.; Huang, L.; Vijh, S.; Johnson, S.; Bonvini, E.; et al. Fc optimization of therapeutic antibodies enhances their ability to kill tumor cells in vitro and controls tumor expansion in vivo via low-affinity activating Fcgamma receptors. Cancer Res. 2007, 67, 8882–8890. [Google Scholar] [CrossRef] [Green Version]
- Umaña, P.; Jean-Mairet, J.; Moudry, R.; Amstutz, H.; Bailey, J.E. Engineered glycoforms of an antineuroblastoma IgG1 with optimized antibody-dependent cellular cytotoxic activity. Nat. Biotechnol. 1999, 17, 176–180. [Google Scholar] [CrossRef]
- Davies, J.; Jiang, L.; Pan, L.Z.; LaBarre, M.J.; Anderson, D.; Reff, M. Expression of GnTIII in a recombinant anti-CD20 CHO production cell line: Expression of antibodies with altered glycoforms leads to an increase in ADCC through higher affinity for FC gamma RIII. Biotechnol. Bioeng. 2001, 74, 288–294. [Google Scholar] [CrossRef] [PubMed]
- Shinkawa, T.; Nakamura, K.; Yamane, N.; Shoji-Hosaka, E.; Kanda, Y.; Sakurada, M.; Uchida, K.; Anazawa, H.; Satoh, M.; Yamasaki, M.; et al. The absence of fucose but not the presence of galactose or bisecting N-acetylglucosamine of human IgG1 complex-type oligosaccharides shows the critical role of enhancing antibody-dependent cellular cytotoxicity. J. Biol. Chem. 2003, 278, 3466–3473. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Schmied, B.J.; Riegg, F.; Zekri, L.; Grosse-Hovest, L.; Bühring, H.-J.; Jung, G.; Salih, H.R. An Fc-Optimized CD133 Antibody for Induction of Natural Killer Cell Reactivity against Colorectal Cancer. Cancers 2019, 11, 789. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Klisovic, R.B.; Leung, W.H.; Brugger, W.; Dirnberger-Hertweck, M.; Winderlich, M.; Ambarkhane, S.V.; Jabbour, E.J. A phase 2a, single-arm, open-label study of tafasitamab, a humanized, Fc-modified, anti-CD19 antibody, in patients with relapsed/refractory B-precursor cell acute lymphoblastic leukemia. Cancer 2021, 127, 4190–4197. [Google Scholar] [CrossRef] [PubMed]
- Sondel, P.M.; Gillies, S.D. Current and Potential Uses of Immunocytokines as Cancer Immunotherapy. Antibodies 2012, 1, 149–171. [Google Scholar] [CrossRef]
- Weiss, T.; Puca, E.; Silginer, M.; Hemmerle, T.; Pazahr, S.; Bink, A.; Weller, M.; Neri, D.; Roth, P. Immunocytokines are a promising immunotherapeutic approach against glioblastoma. Sci. Transl. Med. 2020, 12, eabb2311. [Google Scholar] [CrossRef] [PubMed]
- Gauthier, L.; Morel, A.; Anceriz, N.; Rossi, B.; Blanchard-Alvarez, A.; Grondin, G.; Trichard, S.; Cesari, C.; Sapet, M.; Bosco, F.; et al. Multifunctional Natural Killer Cell Engagers Targeting NKp46 Trigger Protective Tumor Immunity. Cell 2019, 177, 1701–1713.e16. [Google Scholar] [CrossRef]
- Davis, Z.B.; Vallera, D.A.; Miller, J.S.; Felices, M. Natural killer cells unleashed: Checkpoint receptor blockade and BiKE/TriKE utilization in NK-mediated anti-tumor immunotherapy. Semin. Immunol. 2017, 31, 64–75. [Google Scholar] [CrossRef] [PubMed]
- Felices, M.; Lenvik, T.R.; Davis, Z.B.; Miller, J.S.; Vallera, D.A. Generation of BiKEs and TriKEs to Improve NK Cell-Mediated Targeting of Tumor Cells. Methods Mol. Biol. 2016, 1441, 333–346. [Google Scholar] [CrossRef] [Green Version]
- Vallera, D.A.; Felices, M.; McElmurry, R.; McCullar, V.; Zhou, X.; Schmohl, J.U.; Zhang, B.; Lenvik, A.J.; Panoskaltsis-Mortari, A.; Verneris, M.R.; et al. IL15 Trispecific Killer Engagers (TriKE) Make Natural Killer Cells Specific to CD33+ Targets While Also Inducing Persistence, In Vivo Expansion, and Enhanced Function. Clin. Cancer Res. 2016, 22, 3440–3450. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Navin, I.; Lam, M.T.; Parihar, R. Design and Implementation of NK Cell-Based Immunotherapy to Overcome the Solid Tumor Microenvironment. Cancers 2020, 12, 3871. [Google Scholar] [CrossRef] [PubMed]
- Moore, G.L.; Bautista, C.; Pong, E.; Nguyen, D.-H.T.; Jacinto, J.; Eivazi, A.; Muchhal, U.S.; Karki, S.; Chu, S.Y.; Lazar, G.A. A novel bispecific antibody format enables simultaneous bivalent and monovalent co-engagement of distinct target antigens. MAbs 2011, 3, 546–557. [Google Scholar] [CrossRef]
- Holliger, P.; Hudson, P.J. Engineered antibody fragments and the rise of single domains. Nat. Biotechnol. 2005, 23, 1126–1136. [Google Scholar] [CrossRef]
- Reiners, K.S.; Kessler, J.; Sauer, M.; Rothe, A.; Hansen, H.P.; Reusch, U.; Hucke, C.; Köhl, U.; Dürkop, H.; Engert, A.; et al. Rescue of impaired NK cell activity in hodgkin lymphoma with bispecific antibodies in vitro and in patients. Mol. Ther. 2013, 21, 895–903. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Renner, C.; Hartmann, F.; Jung, W.; Deisting, C.; Juwana, M.; Pfreundschuh, M. Initiation of humoral and cellular immune responses in patients with refractory Hodgkin’s disease by treatment with an anti-CD16/CD30 bispecific antibody. Cancer Immunol. Immunother. 2000, 49, 173–180. [Google Scholar] [CrossRef] [PubMed]
- Xie, Z.; Shi, M.; Feng, J.; Yu, M.; Sun, Y.; Shen, B.; Guo, N. A trivalent anti-erbB2/anti-CD16 bispecific antibody retargeting NK cells against human breast cancer cells. Biochem. Biophys. Res. Commun. 2003, 311, 307–312. [Google Scholar] [CrossRef]
- Shahied, L.S.; Tang, Y.; Alpaugh, R.K.; Somer, R.; Greenspon, D.; Weiner, L.M. Bispecific minibodies targeting HER2/neu and CD16 exhibit improved tumor lysis when placed in a divalent tumor antigen binding format. J. Biol. Chem. 2004, 279, 53907–53914. [Google Scholar] [CrossRef] [Green Version]
- Stockmeyer, B.; Valerius, T.; Repp, R.; Heijnen, I.A.; Bühring, H.J.; Deo, Y.M.; Kalden, J.R.; Gramatzki, M.; van de Winkel, J.G. Preclinical studies with Fc(gamma)R bispecific antibodies and granulocyte colony-stimulating factor-primed neutrophils as effector cells against HER-2/neu overexpressing breast cancer. Cancer Res. 1997, 57, 696–701. [Google Scholar] [PubMed]
- Glorius, P.; Baerenwaldt, A.; Kellner, C.; Staudinger, M.; Dechant, M.; Stauch, M.; Beurskens, F.J.; Parren, P.W.H.I.; van de Winkel, J.G.J.; Valerius, T.; et al. The novel tribody [(CD20)(2)xCD16] efficiently triggers effector cell-mediated lysis of malignant B cells. Leukemia 2013, 27, 190–201. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Kellner, C.; Bruenke, J.; Horner, H.; Schubert, J.; Schwenkert, M.; Mentz, K.; Barbin, K.; Stein, C.; Peipp, M.; Stockmeyer, B.; et al. Heterodimeric bispecific antibody-derivatives against CD19 and CD16 induce effective antibody-dependent cellular cytotoxicity against B-lymphoid tumor cells. Cancer Lett. 2011, 303, 128–139. [Google Scholar] [CrossRef]
- Bruenke, J.; Barbin, K.; Kunert, S.; Lang, P.; Pfeiffer, M.; Stieglmaier, K.; Niethammer, D.; Stockmeyer, B.; Peipp, M.; Repp, R.; et al. Effective lysis of lymphoma cells with a stabilised bispecific single-chain Fv antibody against CD19 and FcgammaRIII (CD16). Br. J. Haematol. 2005, 130, 218–228. [Google Scholar] [CrossRef] [PubMed]
- Singer, H.; Kellner, C.; Lanig, H.; Aigner, M.; Stockmeyer, B.; Oduncu, F.; Schwemmlein, M.; Stein, C.; Mentz, K.; Mackensen, A.; et al. Effective elimination of acute myeloid leukemic cells by recombinant bispecific antibody derivatives directed against CD33 and CD16. J. Immunother. 2010, 33, 599–608. [Google Scholar] [CrossRef]
- Kügler, M.; Stein, C.; Kellner, C.; Mentz, K.; Saul, D.; Schwenkert, M.; Schubert, I.; Singer, H.; Oduncu, F.; Stockmeyer, B.; et al. A recombinant trispecific single-chain Fv derivative directed against CD123 and CD33 mediates effective elimination of acute myeloid leukaemia cells by dual targeting. Br. J. Haematol. 2010, 150, 574–586. [Google Scholar] [CrossRef]
- Vallera, D.A.; Zhang, B.; Gleason, M.K.; Oh, S.; Weiner, L.M.; Kaufman, D.S.; McCullar, V.; Miller, J.S.; Verneris, M.R. Heterodimeric bispecific single-chain variable-fragment antibodies against EpCAM and CD16 induce effective antibody-dependent cellular cytotoxicity against human carcinoma cells. Cancer Biother. Radiopharm. 2013, 28, 274–282. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Schmohl, J.U.; Gleason, M.K.; Dougherty, P.R.; Miller, J.S.; Vallera, D.A. Heterodimeric Bispecific Single Chain Variable Fragments (scFv) Killer Engagers (BiKEs) Enhance NK-cell Activity Against CD133+ Colorectal Cancer Cells. Target. Oncol. 2016, 11, 353–361. [Google Scholar] [CrossRef] [Green Version]
- Asano, R.; Nakayama, M.; Kawaguchi, H.; Kubota, T.; Nakanishi, T.; Umetsu, M.; Hayashi, H.; Katayose, Y.; Unno, M.; Kudo, T.; et al. Construction and humanization of a functional bispecific EGFR × CD16 diabody using a refolding system. FEBS J. 2012, 279, 223–233. [Google Scholar] [CrossRef] [PubMed]
- Kuwahara, A.; Nagai, K.; Nakanishi, T.; Kumagai, I.; Asano, R. Functional Domain Order of an Anti-EGFR × Anti-CD16 Bispecific Diabody Involving NK Cell Activation. Int. J. Mol. Sci. 2020, 21, 8914. [Google Scholar] [CrossRef] [PubMed]
- Vallera, D.A.; Ferrone, S.; Kodal, B.; Hinderlie, P.; Bendzick, L.; Ettestad, B.; Hallstrom, C.; Zorko, N.A.; Rao, A.; Fujioka, N.; et al. NK-Cell-Mediated Targeting of Various Solid Tumors Using a B7-H3 Tri-Specific Killer Engager In Vitro and In Vivo. Cancers 2020, 12, 2659. [Google Scholar] [CrossRef] [PubMed]
- Kellner, C.; Gramatzki, M.; Peipp, M. Promoting natural killer cell functions by recombinant immunoligands mimicking an induced self phenotype. Oncoimmunology 2013, 2, e24481. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Jachimowicz, R.D.; Fracasso, G.; Yazaki, P.J.; Power, B.E.; Borchmann, P.; Engert, A.; Hansen, H.P.; Reiners, K.S.; Marie, M.; von Strandmann, E.P.; et al. Induction of in vitro and in vivo NK cell cytotoxicity using high-avidity immunoligands targeting prostate-specific membrane antigen in prostate carcinoma. Mol. Cancer Ther. 2011, 10, 1036–1045. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Larkin, J.; Chiarion-Sileni, V.; Gonzalez, R.; Grob, J.J.; Cowey, C.L.; Lao, C.D.; Schadendorf, D.; Dummer, R.; Smylie, M.; Rutkowski, P.; et al. Combined Nivolumab and Ipilimumab or Monotherapy in Untreated Melanoma. N. Engl. J. Med. 2015, 373, 23–34. [Google Scholar] [CrossRef] [Green Version]
- Xu, X.; Huang, Z.; Zheng, L.; Fan, Y. The efficacy and safety of anti-PD-1/PD-L1 antibodies combined with chemotherapy or CTLA4 antibody as a first-line treatment for advanced lung cancer. Int. J. Cancer 2018, 142, 2344–2354. [Google Scholar] [CrossRef] [Green Version]
- Omuro, A.; Vlahovic, G.; Lim, M.; Sahebjam, S.; Baehring, J.; Cloughesy, T.; Voloschin, A.; Ramkissoon, S.H.; Ligon, K.L.; Latek, R.; et al. Nivolumab with or without ipilimumab in patients with recurrent glioblastoma: Results from exploratory phase I cohorts of CheckMate 143. Neuro-Oncol. 2018, 20, 674–686. [Google Scholar] [CrossRef]
- Reardon, D.A.; Brandes, A.A.; Omuro, A.; Mulholland, P.; Lim, M.; Wick, A.; Baehring, J.; Ahluwalia, M.S.; Roth, P.; Bähr, O.; et al. Effect of Nivolumab vs Bevacizumab in Patients with Recurrent Glioblastoma: The CheckMate 143 Phase 3 Randomized Clinical Trial. JAMA Oncol. 2020, 6, 1003–1010. [Google Scholar] [CrossRef]
- Shevtsov, M.; Pitkin, E.; Ischenko, A.; Stangl, S.; Khachatryan, W.; Galibin, O.; Edmond, S.; Lobinger, D.; Multhoff, G. Ex vivo Hsp70-Activated NK Cells in Combination with PD-1 Inhibition Significantly Increase Overall Survival in Preclinical Models of Glioblastoma and Lung Cancer. Front. Immunol. 2019, 10, 454. [Google Scholar] [CrossRef] [PubMed]
- André, P.; Denis, C.; Soulas, C.; Bourbon-Caillet, C.; Lopez, J.; Arnoux, T.; Bléry, M.; Bonnafous, C.; Gauthier, L.; Morel, A.; et al. Anti-NKG2A mAb Is a Checkpoint Inhibitor that Promotes Anti-tumor Immunity by Unleashing Both T and NK Cells. Cell 2018, 175, 1731–1743.e13. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Romagné, F.; André, P.; Spee, P.; Zahn, S.; Anfossi, N.; Gauthier, L.; Capanni, M.; Ruggeri, L.; Benson, D.M., Jr.; Blaser, B.W.; 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 2009, 114, 2667–2677. [Google Scholar] [CrossRef] [PubMed]
- Benson, D.M., Jr.; Hofmeister, C.C.; Padmanabhan, S.; Suvannasankha, A.; Jagannath, S.; Abonour, R.; Bakan, C.; Andre, P.; Efebera, Y.; Tiollier, J.; et al. A phase 1 trial of the anti-KIR antibody IPH2101 in patients with relapsed/refractory multiple myeloma. Blood 2012, 120, 4324–4333. [Google Scholar] [CrossRef] [PubMed]
- Rupp, L.J.; Schumann, K.; Roybal, K.T.; Gate, R.E.; Ye, C.J.; Lim, W.A.; Marson, A. CRISPR/Cas9-mediated PD-1 disruption enhances anti-tumor efficacy of human chimeric antigen receptor T cells. Sci. Rep. 2017, 7, 737. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Zhang, C.; Peng, Y.; Hublitz, P.; Zhang, H.; Dong, T. Genetic abrogation of immune checkpoints in antigen-specific cytotoxic T-lymphocyte as a potential alternative to blockade immunotherapy. Sci. Rep. 2018, 8, 5549. [Google Scholar] [CrossRef] [PubMed]
- Su, S.; Hu, B.; Shao, J.; Shen, B.; Du, J.; Du, Y.; Zhou, J.; Yu, L.; Zhang, L.; Chen, F.; et al. CRISPR-Cas9 mediated efficient PD-1 disruption on human primary T cells from cancer patients. Sci. Rep. 2016, 6, 20070. [Google Scholar] [CrossRef] [Green Version]
- Lu, Y.; Xue, J.; Deng, T.; Zhou, X.; Yu, K.; Deng, L.; Huang, M.; Yi, X.; Liang, M.; Wang, Y.; et al. Safety and feasibility of CRISPR-edited T cells in patients with refractory non-small-cell lung cancer. Nat. Med. 2020, 26, 732–740. [Google Scholar] [CrossRef] [PubMed]
- Stadtmauer, E.A.; Fraietta, J.A.; Davis, M.M.; Cohen, A.D.; Weber, K.L.; Lancaster, E.; Mangan, P.A.; Kulikovskaya, I.; Gupta, M.; Chen, F.; et al. CRISPR-engineered T cells in patients with refractory cancer. Science 2020, 367. [Google Scholar] [CrossRef] [PubMed]
- Huang, R.-S.; Lai, M.-C.; Shih, H.-A.; Lin, S. A robust platform for expansion and genome editing of primary human natural killer cells. J. Exp. Med. 2021, 218, e20201529. [Google Scholar] [CrossRef] [PubMed]
- Morimoto, T.; Nakazawa, T.; Matsuda, R.; Nishimura, F.; Nakamura, M.; Yamada, S.; Nakagawa, I.; Park, Y.-S.; Tsujimura, T.; Nakase, H. CRISPR-Cas9–Mediated TIM3 Knockout in Human Natural Killer Cells Enhances Growth Inhibitory Effects on Human Glioma Cells. Int. J. Mol. Sci. 2021, 22, 3489. [Google Scholar] [CrossRef]
- Guo, X.; Mahlakõiv, T.; Ye, Q.; Somanchi, S.; He, S.; Rana, H.; DiFiglia, A.; Gleason, J.; van der Touw, W.; Hariri, R.; et al. CBLB ablation with CRISPR/Cas9 enhances cytotoxicity of human placental stem cell-derived NK cells for cancer immunotherapy. J. Immunother. Cancer 2021, 9, e001975. [Google Scholar] [CrossRef]
- Pomeroy, E.J.; Hunzeker, J.T.; Kluesner, M.G.; Lahr, W.S.; Smeester, B.A.; Crosby, M.R.; Lonetree, C.; Yamamoto, K.; Bendzick, L.; Miller, J.S.; et al. A Genetically Engineered Primary Human Natural Killer Cell Platform for Cancer Immunotherapy. Mol. Ther. 2020, 28, 52–63. [Google Scholar] [CrossRef]
- Delconte, R.B.; Kolesnik, T.B.; Dagley, L.F.; Rautela, J.; Shi, W.; Putz, E.M.; Stannard, K.; Zhang, J.-G.; Teh, C.; Firth, M.; et al. CIS is a potent checkpoint in NK cell-mediated tumor immunity. Nat. Immunol. 2016, 17, 816–824. [Google Scholar] [CrossRef] [PubMed]
- Rautela, J.; Surgenor, E.; Huntington, N.D. Efficient genome editing of human natural killer cells by CRISPR RNP. bioRxiv 2018, 406934. [Google Scholar] [CrossRef]
- Sutlu, T.; Nyström, S.; Gilljam, M.; Stellan, B.; Applequist, S.E.; Alici, E. Inhibition of intracellular antiviral defense mechanisms augments lentiviral transduction of human natural killer cells: Implications for gene therapy. Hum. Gene Ther. 2012, 23, 1090–1100. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Rautela, J.; Surgenor, E.; Huntington, N.D. Drug target validation in primary human natural killer cells using CRISPR RNP. J. Leukoc. Biol. 2020, 108, 1397–1408. [Google Scholar] [CrossRef]
- Raedler, L. Velcade (Bortezomib) Receives 2 New FDA Indications: For Retreatment of Patients with Multiple Myeloma and for First-Line Treatment of Patients with Mantle-Cell Lymphoma. Am. Health Drug Benefits 2015, 8, 135–140. [Google Scholar] [PubMed]
- Luna, J.I.; Grossenbacher, S.K.; Sturgill, I.R.; Ames, E.; Judge, S.J.; Bouzid, L.A.; Darrow, M.A.; Murphy, W.J.; Canter, R.J. Bortezomib Augments Natural Killer Cell Targeting of Stem-Like Tumor Cells. Cancers 2019, 11, 85. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Höring, E.; Podlech, O.; Silkenstedt, B.; Rota, I.A.; Adamopoulou, E.; Naumann, U. The Histone Deacetylase Inhibitor Trichostatin A Promotes Apoptosis and Antitumor Immunity in Glioblastoma Cells. Anticancer Res. 2013, 33, 1351–1360. [Google Scholar] [PubMed]
- Chitadze, G.; Lettau, M.; Luecke, S.; Wang, T.; Janssen, O.; Fürst, D.; Mytilineos, J.; Wesch, D.; Oberg, H.-H.; Held-Feindt, J.; et al. NKG2D- and T-cell receptor-dependent lysis of malignant glioma cell lines by human γδ T cells: Modulation by temozolomide and A disintegrin and metalloproteases 10 and 17 inhibitors. Oncoimmunology 2016, 5, e1093276. [Google Scholar] [CrossRef] [PubMed]
Receptor | Ligand(s) | Function | Reference |
---|---|---|---|
NKp30 | B7-H6, BAT3, viral ligands, heparan sulfate proteoglycan | Stimulatory | [42,43] |
NKp44 | Viral ligands, heparan sulfate proteoglycan, nidogen-1, PCNA | Stimulatory | [42,43] |
NKp46 | Viral ligands, heparan sulfate proteoglycan, vimentin | Stimulatory | [42,43] |
FcγRIIIa | Fc portion of IgG | Stimulatory | [44] |
DNAM-1 | PVR (CD155), nectin-2 (CD112) | Co-stimulatory | [45] |
CD94/NKG2A | HLA-E | Inhibitory | [46] |
CD94/NKG2C, E | HLA-E | Stimulatory | [47] |
NKG2D | ULBP1-6, MIC-A, B | Stimulatory | [48] |
KIR2DS(1-5)/KIR3DS1 | MHC class I | Stimulatory | [49] |
KIR2DL(1-5)/KIR3DL(1-3) | MHC class I | Inhibitory | [49] |
2B4 | CD48 | Co-stimulatory/inhibitory | [50] |
PD-1 | PD-L1, PD-L2 | Inhibitory | [51] |
CTLA-4 | CD28 | Inhibitory | [51] |
TIGIT | PVR (CD155), nectin-2 (CD112), nectin-3 (CD113), nectin-4 | Inhibitory | [52,53] |
LAG-3 | HLA class II, galectin-3 | Inhibitory | [51,54] |
TIM-3 | Phosphatidylserine, HMGB1, CEACAM1 glycoprotein, galectin-9 | Co-inhibitory | [51,55] |
Tactile (CD96) | PVR (CD155), nectin-1 (CD111) | Co-inhibitory/stimulatory | [56] |
Method | Markers | Infiltration in GBM | Main Phenotypic and Functional Alterations | Sample Size | Reference |
---|---|---|---|---|---|
Flow cytometry | CD14− CD3− CD56+ | 2.11 ± 0.54% of leucocytes | Predominance of CD56dim CD16− NK cells. Some NK cells lack expression of NKG2D (42.55%) | n = 8 | [128] |
Flow cytometry | CD19− CD3− CD56+ CD16high, among others | 1 ± 5% of lymphocytes and 0.05 ± 0.05% of all cells in the tumor sample | Not measured | n = 53 | [129] |
Flow cytometry | CD45+ CD3− NKp46+ | Not measured | ↓ Activating receptors’ protein levels compared to PB NK cells (NKp30, NKG2D, DNAM1, and CD2); ↑ CD69, a marker of activation; ↑ CD9, a TGF-β-induced molecule, described previously as being related to impaired activation of NK by modulation of NKG2D levels | n = 8 for NKp30, CD69, NKG2D and CD2) n = 9 for DNAM-1 n = 5 for CD9 | [133] |
Mass cytometry (CyTOF) | CD3− CD56+ CD16+ | ~10% of leucocytes, with no significant difference from PB NK cells. | ↑ CXCR3 protein and ↓ IFN-γ transcripts compared to PB NK cells | n = 8 | [134] |
RNA-Seq - CIBERSORT (In silico analysis) | 13 genes’ expression profiles | 9.8% of deconvoluted leukocytes | More quiescent than activated NK cells | n = 540 | [132] |
Immunohistochemistry | CD56+ | ~50% of samples showed a rare level of GBM intratumoral CD56+ cells (<5 NK cells in 10 high-power fields (HPFs)) The lasting % of samples showed focal (2–20 cells/HPF) or median (20–100 cells/HPF) infiltration, but not extensive infiltration (>100 cell/HPF) in intratumoral tissue; 49% of samples showed some level of perivascular NK infiltration, and 89% of samples showed some intratumoral infiltration | Not measured | n = 63 | [127] |
Publisher’s Note: MDPI stays neutral with regard to jurisdictional claims in published maps and institutional affiliations. |
© 2022 by the authors. Licensee MDPI, Basel, Switzerland. This article is an open access article distributed under the terms and conditions of the Creative Commons Attribution (CC BY) license (https://creativecommons.org/licenses/by/4.0/).
Share and Cite
da Silva, L.H.R.; Catharino, L.C.C.; da Silva, V.J.; Evangelista, G.C.M.; Barbuto, J.A.M. The War Is on: The Immune System against Glioblastoma—How Can NK Cells Drive This Battle? Biomedicines 2022, 10, 400. https://doi.org/10.3390/biomedicines10020400
da Silva LHR, Catharino LCC, da Silva VJ, Evangelista GCM, Barbuto JAM. The War Is on: The Immune System against Glioblastoma—How Can NK Cells Drive This Battle? Biomedicines. 2022; 10(2):400. https://doi.org/10.3390/biomedicines10020400
Chicago/Turabian Styleda Silva, Lucas Henrique Rodrigues, Luana Correia Croda Catharino, Viviane Jennifer da Silva, Gabriela Coeli Menezes Evangelista, and José Alexandre Marzagão Barbuto. 2022. "The War Is on: The Immune System against Glioblastoma—How Can NK Cells Drive This Battle?" Biomedicines 10, no. 2: 400. https://doi.org/10.3390/biomedicines10020400
APA Styleda Silva, L. H. R., Catharino, L. C. C., da Silva, V. J., Evangelista, G. C. M., & Barbuto, J. A. M. (2022). The War Is on: The Immune System against Glioblastoma—How Can NK Cells Drive This Battle? Biomedicines, 10(2), 400. https://doi.org/10.3390/biomedicines10020400