Recent Advances to Augment NK Cell Cancer Immunotherapy Using Nanoparticles
Abstract
1. Introduction
2. NK Cell Cancer Immunotherapy Clinical Trials
3. Challenges of NK Cell Cancer Immunotherapy
4. Nanomaterials for NK Cell Cancer-Immunotherapy
4.1. Nanoparticle-Mediated Conversion of Immune Suppressive TME for NK Cell Cancer Immunotherapy
4.2. Nanoparticle-Mediated NK Cell Homing and Infiltration
4.3. Nanoparticle-Mediated NK Cell Receptor-Ligand Activation and Nano-Engagers
5. Conclusions and Future Outlook
Author Contributions
Funding
Institutional Review Board Statement
Informed Consent Statement
Data Availability Statement
Acknowledgments
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. [Google Scholar] [CrossRef] [PubMed]
- Mellman, I.; Coukos, G.; Dranoff, G. Cancer immunotherapy comes of age. Nature 2011, 480, 480–489. [Google Scholar] [CrossRef]
- Wei, S.C.; Duffy, C.R.; Allison, J.P. Fundamental Mechanisms of Immune Checkpoint Blockade Therapy. Cancer Discov. 2018, 8, 1069–1086. [Google Scholar] [CrossRef]
- June, C.H.; Sadelain, M. Chimeric Antigen Receptor Therapy. N. Engl. J. Med. 2018, 379, 64–73. [Google Scholar] [CrossRef]
- Waldmann, T.A. Cytokines in Cancer Immunotherapy. Cold Spring Harb. Perspect. Biol. 2018, 10. [Google Scholar] [CrossRef]
- Bald, T.; Krummel, M.F.; Smyth, M.J.; Barry, K.C. The NK cell-cancer cycle: Advances and new challenges in NK cell-based immunotherapies. Nat. Immunol. 2020, 21, 835–847. [Google Scholar] [CrossRef]
- Banchereau, J.; Palucka, K. Immunotherapy: Cancer vaccines on the move. Nat. Rev. Clin. Oncol. 2018, 15, 9–10. [Google Scholar] [CrossRef]
- Farkona, S.; Diamandis, E.P.; Blasutig, I.M. Cancer immunotherapy: The beginning of the end of cancer? BMC Med. 2016, 14, 73. [Google Scholar] [CrossRef]
- Falzone, L.; Salomone, S.; Libra, M. Evolution of Cancer Pharmacological Treatments at the Turn of the Third Millennium. Front. Pharmacol. 2018, 9, 1300. [Google Scholar] [CrossRef] [PubMed]
- Fares, C.M.; Van Allen, E.M.; Drake, C.G.; Allison, J.P.; Hu-Lieskovan, S. Mechanisms of Resistance to Immune Checkpoint Blockade: Why Does Checkpoint Inhibitor Immunotherapy Not Work for All Patients? Am. Soc. Clin. Oncol. Educ. Book 2019, 39, 147–164. [Google Scholar] [CrossRef] [PubMed]
- Jenkins, R.W.; Barbie, D.A.; Flaherty, K.T. Mechanisms of resistance to immune checkpoint inhibitors. Br. J. Cancer 2018, 118, 9–16. [Google Scholar] [CrossRef] [PubMed]
- Alsaab, H.O.; Sau, S.; Alzhrani, R.; Tatiparti, K.; Bhise, K.; Kashaw, S.K.; Iyer, A.K. PD-1 and PD-L1 Checkpoint Signaling Inhibition for Cancer Immunotherapy: Mechanism, Combinations, and Clinical Outcome. Front. Pharmacol. 2017, 8, 561. [Google Scholar] [CrossRef] [PubMed]
- Riley, R.S.; June, C.H.; Langer, R.; Mitchell, M.J. Delivery technologies for cancer immunotherapy. Nat. Rev. Drug Discov. 2019, 18, 175–196. [Google Scholar] [CrossRef] [PubMed]
- Ljunggren, H.G.; Malmberg, K.J. Prospects for the use of NK cells in immunotherapy of human cancer. Nat. Rev. Immunol. 2007, 7, 329–339. [Google Scholar] [CrossRef]
- Parkhurst, M.R.; Riley, J.P.; Dudley, M.E.; Rosenberg, S.A. 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]
- 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]
- Vivier, E.; Artis, D.; Colonna, M.; Diefenbach, A.; Di Santo, J.P.; Eberl, G.; Koyasu, S.; Locksley, R.M.; McKenzie, A.N.J.; Mebius, R.E.; et al. Innate Lymphoid Cells: 10 Years On. Cell 2018, 174, 1054–1066. [Google Scholar] [CrossRef]
- Huntington, N.D.; Cursons, J.; Rautela, J. The cancer-natural killer cell immunity cycle. Nat. Rev. Cancer 2020, 20, 437–454. [Google Scholar] [CrossRef] [PubMed]
- Farag, S.S.; Caligiuri, M.A. Human natural killer cell development and biology. Blood Rev. 2006, 20, 123–137. [Google Scholar] [CrossRef]
- Lanier, L.L. Natural killer cell receptor signaling. Curr. Opin. Immunol. 2003, 15, 308–314. [Google Scholar] [CrossRef]
- Orr, M.T.; Lanier, L.L. Natural Killer Cell Education and Tolerance. Cell 2010, 142, 847–856. [Google Scholar] [CrossRef] [PubMed]
- Wensveen, F.M.; Jelencic, V.; Polic, B. NKG2D: A Master Regulator of Immune Cell Responsiveness. Front. Immunol. 2018, 9, 441. [Google Scholar] [CrossRef] [PubMed]
- Liu, S.; Galat, V.; Galat, Y.; Lee, Y.K.A.; Wainwright, D.; Wu, J. NK cell-based cancer immunotherapy: From basic biology to clinical development. J. Hematol. Oncol. 2021, 14, 7. [Google Scholar] [CrossRef] [PubMed]
- Romee, R.; Rosario, M.; Berrien-Elliott, M.M.; Wagner, J.A.; Jewell, B.A.; Schappe, T.; Leong, J.W.; Abdel-Latif, S.; Schneider, S.E.; Willey, S.; et al. Cytokine-induced memory-like natural killer cells exhibit enhanced responses against myeloid leukemia. Sci. Transl. Med. 2016, 8, 357ra123. [Google Scholar] [CrossRef]
- Bachanova, V.; Sarhan, D.; DeFor, T.E.; Cooley, S.; Panoskaltsis-Mortari, A.; Blazar, B.R.; Curtsinger, J.M.; Burns, L.; Weisdorf, D.J.; Miller, J.S. Haploidentical natural killer cells induce remissions in non-Hodgkin lymphoma patients with low levels of immune-suppressor cells. Cancer Immunol. Immunother. 2018, 67, 483–494. [Google Scholar] [CrossRef]
- 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]
- Nayyar, G.; Chu, Y.; Cairo, M.S. Overcoming Resistance to Natural Killer Cell Based Immunotherapies for Solid Tumors. Front. Oncol. 2019, 9, 51. [Google Scholar] [CrossRef] [PubMed]
- Muller, T.; Uherek, C.; Maki, G.; Chow, K.U.; Schimpf, A.; Klingemann, H.G.; Tonn, T.; Wels, W.S. Expression of a CD20-specific chimeric antigen receptor enhances cytotoxic activity of NK cells and overcomes NK-resistance of lymphoma and leukemia cells. Cancer Immunol. Immunother. 2008, 57, 411–423. [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]
- Cerwenka, A.; Lanier, L.L. Natural killer cell memory in infection, inflammation and cancer. Nat. Rev. Immunol. 2016, 16, 112. [Google Scholar] [CrossRef]
- Fehniger, T.A.; Cooper, M.A. Harnessing NK Cell Memory for Cancer Immunotherapy. Trends Immunol. 2016, 37, 877–888. [Google Scholar] [CrossRef]
- De Lazaro, I.; Mooney, D.J. A nanoparticle’s pathway into tumours. Nat. Mater. 2020, 19, 486–487. [Google Scholar] [CrossRef]
- Nam, J.; Son, S.; Park, K.S.; Zou, W.; Shea, L.D.; Moon, J.J. Cancer nanomedicine for combination cancer immunotherapy. Nat. Rev. Mater. 2019, 4, 398–414. [Google Scholar] [CrossRef]
- Irvine, D.J.; Dane, E.L. Enhancing cancer immunotherapy with nanomedicine. Nat. Rev. Immunol. 2020, 20, 321–334. [Google Scholar] [CrossRef] [PubMed]
- Phung, C.D.; Tran, T.H.; Kim, J.O. Engineered nanoparticles to enhance natural killer cell activity towards onco-immunotherapy: A review. Arch. Pharmacal. Res. 2020, 43, 32–45. [Google Scholar] [CrossRef]
- 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]
- Curti, A.; Ruggeri, L.; D’Addio, A.; Bontadini, A.; Dan, E.; Motta, M.R.; Trabanelli, S.; Giudice, V.; Urbani, E.; Martinelli, G.; et al. Successful transfer of alloreactive haploidentical KIR ligand-mismatched natural killer cells after infusion in elderly high risk acute myeloid leukemia patients. Blood 2011, 118, 3273–3279. [Google Scholar] [CrossRef] [PubMed]
- 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] [PubMed]
- Krause, S.W.; 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]
- Geller, M.A.; Cooley, S.; Judson, P.L.; Ghebre, R.; Carson, L.F.; Argenta, P.A.; Jonson, A.L.; Panoskaltsis-Mortari, A.; Curtsinger, J.; McKenna, D.; et al. A phase II study of allogeneic natural killer cell therapy to treat patients with recurrent ovarian and breast cancer. Cytotherapy 2011, 13, 98–107. [Google Scholar] [CrossRef]
- 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]
- Ishikawa, T.; Okayama, T.; Sakamoto, N.; Ideno, M.; Oka, K.; Enoki, T.; Mineno, J.; Yoshida, N.; Katada, K.; Kamada, K.; et al. Phase I clinical trial of adoptive transfer of expanded natural killer cells in combination with IgG1 antibody in patients with gastric or colorectal cancer. Int. J. Cancer 2018, 142, 2599–2609. [Google Scholar] [CrossRef]
- Oh, S.; Lee, J.H.; Kwack, K.; Choi, S.W. Natural Killer Cell Therapy: A New Treatment Paradigm for Solid Tumors. Cancers 2019, 11, 1534. [Google Scholar] [CrossRef] [PubMed]
- Fang, F.; Xiao, W.; Tian, Z. Challenges of NK cell-based immunotherapy in the new era. Front. Med. 2018, 12, 440–450. [Google Scholar] [CrossRef] [PubMed]
- Domogala, A.; Madrigal, J.A.; Saudemont, A. Natural Killer Cell Immunotherapy: From Bench to Bedside. Front. Immunol. 2015, 6, 264. [Google Scholar] [CrossRef] [PubMed]
- Melaiu, O.; Lucarini, V.; Cifaldi, L.; Fruci, D. Influence of the Tumor Microenvironment on NK Cell Function in Solid Tumors. Front. Immunol. 2019, 10, 3038. [Google Scholar] [CrossRef] [PubMed]
- Abel, A.M.; Yang, C.; Thakar, M.S.; Malarkannan, S. Natural Killer Cells: Development, Maturation, and Clinical Utilization. Front. Immunol. 2018, 9, 1869. [Google Scholar] [CrossRef] [PubMed]
- Melero, I.; Rouzaut, A.; Motz, G.T.; Coukos, G. T-cell and NK-cell infiltration into solid tumors: A key limiting factor for efficacious cancer immunotherapy. Cancer Discov. 2014, 4, 522–526. [Google Scholar] [CrossRef]
- Vitale, M.; Cantoni, C.; Pietra, G.; Mingari, M.C.; Moretta, L. Effect of tumor cells and tumor microenvironment on NK-cell function. Eur. J. Immunol. 2014, 44, 1582–1592. [Google Scholar] [CrossRef]
- Melder, R.J.; Koenig, G.C.; Witwer, B.P.; Safabakhsh, N.; Munn, L.L.; Jain, R.K. During angiogenesis, vascular endothelial growth factor and basic fibroblast growth factor regulate natural killer cell adhesion to tumor endothelium. Nat. Med. 1996, 2, 992–997. [Google Scholar] [CrossRef]
- Myers, J.A.; Miller, J.S. Exploring the NK cell platform for cancer immunotherapy. Nat. Rev. Clin. Oncol. 2021, 18, 85–100. [Google Scholar] [CrossRef]
- Susek, K.H.; Karvouni, M.; Alici, E.; Lundqvist, A. The Role of CXC Chemokine Receptors 1-4 on Immune Cells in the Tumor Microenvironment. Front. Immunol. 2018, 9, 2159. [Google Scholar] [CrossRef]
- Li, F.; Sheng, Y.; Hou, W.; Sampath, P.; Byrd, D.; Thorne, S.; Zhang, Y. CCL5-armed oncolytic virus augments CCR5-engineered NK cell infiltration and antitumor efficiency. J. Immunother. Cancer 2020, 8. [Google Scholar] [CrossRef]
- Alkins, R.; Burgess, A.; Kerbel, R.; Wels, W.S.; Hynynen, K. Early treatment of HER2-amplified brain tumors with targeted NK-92 cells and focused ultrasound improves survival. Neuro Oncol. 2016, 18, 974–981. [Google Scholar] [CrossRef]
- Wu, L.; Zhang, F.; Wei, Z.; Li, X.; Zhao, H.; Lv, H.; Ge, R.; Ma, H.; Zhang, H.; Yang, B.; et al. Magnetic delivery of Fe3O4@polydopamine nanoparticle-loaded natural killer cells suggest a promising anticancer treatment. Biomater. Sci. 2018, 6, 2714–2725. [Google Scholar] [CrossRef]
- Su, Z.; Wang, X.; Zheng, L.; Lyu, T.; Figini, M.; Wang, B.; Procissi, D.; Shangguan, J.; Sun, C.; Pan, L.; et al. MRI-guided interventional natural killer cell delivery for liver tumor treatment. Cancer Med. 2018, 7, 1860–1869. [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]
- Gleason, M.K.; Ross, J.A.; Warlick, E.D.; Lund, T.C.; Verneris, M.R.; Wiernik, A.; Spellman, S.; Haagenson, M.D.; Lenvik, A.J.; Litzow, M.R.; et al. CD16xCD33 bispecific killer cell engager (BiKE) activates NK cells against primary MDS and MDSC CD33+ targets. Blood 2014, 123, 3016–3026. [Google Scholar] [CrossRef] [PubMed]
- Brudno, J.N.; Kochenderfer, J.N. Recent advances in CAR T-cell toxicity: Mechanisms, manifestations and management. Blood Rev. 2019, 34, 45–55. [Google Scholar] [CrossRef]
- Choi, B.; Choi, H.; Yu, B.; Kim, D.-H. Synergistic Local Combination of Radiation and Anti-Programmed Death Ligand 1 Immunotherapy Using Radiation-Responsive Splintery Metallic Nanocarriers. ACS Nano 2020. [Google Scholar] [CrossRef] [PubMed]
- Kim, D.H.; Guo, Y.; Zhang, Z.; Procissi, D.; Nicolai, J.; Omary, R.A.; Larson, A.C. Temperature-Sensitive Magnetic Drug Carriers for Concurrent Gemcitabine Chemohyperthermia. Adv. Healthc. Mater. 2014, 3, 714–724. [Google Scholar] [CrossRef]
- Yu, B.; Choi, B.S.; Li, W.G.; Kim, D.H. Magnetic field boosted ferroptosis-like cell death and responsive MRI using hybrid vesicles for cancer immunotherapy. Nat. Commun. 2020, 11. [Google Scholar] [CrossRef]
- Kim, D.H.; Larson, A.C. Deoxycholate bile acid directed synthesis of branched Au nanostructures for near infrared photothermal ablation. Biomaterials 2015, 56, 154–164. [Google Scholar] [CrossRef] [PubMed]
- Xu, Z.; Wang, Y.; Zhang, L.; Huang, L. Nanoparticle-Delivered Transforming Growth Factor-β siRNA Enhances Vaccination against Advanced Melanoma by Modifying Tumor Microenvironment. ACS Nano 2014, 8, 3636–3645. [Google Scholar] [CrossRef]
- Park, J.; Wrzesinski, S.H.; Stern, E.; Look, M.; Criscione, J.; Ragheb, R.; Jay, S.M.; Demento, S.L.; Agawu, A.; Licona Limon, P.; et al. Combination delivery of TGF-beta inhibitor and IL-2 by nanoscale liposomal polymeric gels enhances tumour immunotherapy. Nat. Mater. 2012, 11, 895–905. [Google Scholar] [CrossRef]
- Fauriat, C.; Long, E.O.; Ljunggren, H.G.; Bryceson, Y.T. Regulation of human NK-cell cytokine and chemokine production by target cell recognition. Blood 2010, 115, 2167–2176. [Google Scholar] [CrossRef] [PubMed]
- Tan, L.; Han, S.; Ding, S.; Xiao, W.; Ding, Y.; Qian, L.; Wang, C.; Gong, W. Chitosan nanoparticle-based delivery of fused NKG2D-IL-21 gene suppresses colon cancer growth in mice. Int. J. Nanomed. 2017, 12, 3095–3107. [Google Scholar] [CrossRef] [PubMed]
- Meraz, I.M.; Majidi, M.; Cao, X.; Lin, H.; Li, L.; Wang, J.; Baladandayuthapani, V.; Rice, D.; Sepesi, B.; Ji, L.; et al. TUSC2 Immunogene Therapy Synergizes with Anti-PD-1 through Enhanced Proliferation and Infiltration of Natural Killer Cells in Syngeneic Kras-Mutant Mouse Lung Cancer Models. Cancer Immunol. Res. 2018, 6, 163–177. [Google Scholar] [CrossRef] [PubMed]
- Atukorale, P.U.; Raghunathan, S.P.; Raguveer, V.; Moon, T.J.; Zheng, C.; Bielecki, P.A.; Wiese, M.L.; Goldberg, A.L.; Covarrubias, G.; Hoimes, C.J.; et al. Nanoparticle Encapsulation of Synergistic Immune Agonists Enables Systemic Codelivery to Tumor Sites and IFNbeta-Driven Antitumor Immunity. Cancer Res. 2019, 79, 5394–5406. [Google Scholar] [CrossRef] [PubMed]
- Park, W.; Gordon, A.C.; Cho, S.; Huang, X.; Harris, K.R.; Larson, A.C.; Kim, D.H. Immunomodulatory Magnetic Microspheres for Augmenting Tumor-Specific Infiltration of Natural Killer (NK) Cells. ACS Appl. Mater. Interfaces 2017, 9, 13819–13824. [Google Scholar] [CrossRef]
- Jang, E.S.; Shin, J.H.; Ren, G.; Park, M.J.; Cheng, K.; Chen, X.; Wu, J.C.; Sunwoo, J.B.; Cheng, Z. The manipulation of natural killer cells to target tumor sites using magnetic nanoparticles. Biomaterials 2012, 33, 5584–5592. [Google Scholar] [CrossRef] [PubMed]
- Chandrasekaran, S.; Chan, M.F.; Li, J.; King, M.R. Super natural killer cells that target metastases in the tumor draining lymph nodes. Biomaterials 2016, 77, 66–76. [Google Scholar] [CrossRef]
- Kim, K.S.; Han, J.H.; Choi, S.H.; Jung, H.Y.; Park, J.D.; An, H.J.; Kim, S.E.; Kim, D.H.; Doh, J.; Han, D.K.; et al. Cationic Nanoparticle-Mediated Activation of Natural Killer Cells for Effective Cancer Immunotherapy. ACS Appl. Mater. Interfaces 2020, 12, 56731–56740. [Google Scholar] [CrossRef] [PubMed]
- Zheng, C.; Wang, Q.; Wang, Y.; Zhao, X.; Gao, K.; Liu, Q.; Zhao, Y.; Zhang, Z.; Zheng, Y.; Cao, J.; et al. In Situ Modification of the Tumor Cell Surface with Immunomodulating Nanoparticles for Effective Suppression of Tumor Growth in Mice. Adv. Mater. 2019, 31, e1902542. [Google Scholar] [CrossRef]
- Au, K.M.; Park, S.I.; Wang, A.Z. Trispecific natural killer cell nanoengagers for targeted chemoimmunotherapy. Sci. Adv. 2020, 6, eaba8564. [Google Scholar] [CrossRef] [PubMed]
- Liu, C.; Lai, H.; Chen, T. Boosting Natural Killer Cell-Based Cancer Immunotherapy with Selenocystine/Transforming Growth Factor-Beta Inhibitor-Encapsulated Nanoemulsion. ACS Nano 2020, 14, 11067–11082. [Google Scholar] [CrossRef]
- Zhang, Y.; Li, N.; Suh, H.; Irvine, D.J. Nanoparticle anchoring targets immune agonists to tumors enabling anti-cancer immunity without systemic toxicity. Nat. Commun. 2018, 9, 6. [Google Scholar] [CrossRef]
- Frick, S.U.; Domogalla, M.P.; Baier, G.; Wurm, F.R.; Mailander, V.; Landfester, K.; Steinbrink, K. Interleukin-2 Functionalized Nanocapsules for T Cell-Based Immunotherapy. ACS Nano 2016, 10, 9216–9226. [Google Scholar] [CrossRef]
- Yao, H.; Ng, S.S.; Huo, L.F.; Chow, B.K.; Shen, Z.; Yang, M.; Sze, J.; Ko, O.; Li, M.; Yue, A.; et al. Effective Melanoma Immunotherapy with Interleukin-2 Delivered by a Novel Polymeric Nanoparticle. Mol. Cancer Ther. 2011, 10, 1082–1092. [Google Scholar] [CrossRef] [PubMed]
- Paust, S.; Senman, B.; von Andrian, U.H. Adaptive immune responses mediated by natural killer cells. Immunol. Rev. 2010, 235, 286–296. [Google Scholar] [CrossRef]
Year | Cell Source | Pre-Treatment | Tumor Type | Combination | Ref. |
---|---|---|---|---|---|
2008 | Autologous | IL-2 | CML, Pancreatic cancer, Colorectal cancer, Multiple myeloma, Non-small cell lung cancer | Bortezomib | NCT00720785 |
2012 | UCB Allogenic | Multiple Myeloma | Elotuzumab, Lenalidomide, Melphalan | NCT01729091 | |
2013 | PBMC Allogenic | IL-12, IL-15, IL-18 | AML, MDS | ALT-803. | NCT01898793 |
2013 | PBMC Allogenic | Neuroblastoma | Anti-GD2 | NCT01857934 | |
2014 | Autologous | IL-2 | HER2+ Breast and Gastric Cancer | Trastuzumab | NCT02030561 |
2015 | Autologous | IL-2 | Head and Neck Cancer | Cetuximab | NCT02507154 |
2016 | PBMC Allogenic | IL-2 | Neuroblastoma | Anti-GD2 | NCT02650648 |
2016 | PBMC Allogenic | Hematologic, solid cancers | ALT803 | NCT02890758 | |
2016 | Autologous | IL-15 | Multiple Myeloma | Elotuzumab | NCT03003728 |
2017 | PBMC Allogenic | IL-15, GSK3beta inhibitor | Advanced solid tumors | Trastuzumab, Cetuximab | NCT03319459 |
2017 | PBMC Allogenic | IL-15, GSK3beta inhibitor | Ovarian cancer | IL-2 | NCT03213964 |
2017 | UCB Allogenic | NHL | Rituximab | NCT03019640 | |
2017 | PBMC Allogenic | IL-2 | Neuroblastoma Recurrent | Anti-GD2 | NCT03242603 |
2018 | UCB Allogenic | Relapsed or Refractory Solid Tumors | Cyclophosphamide, Etoposide | NCT03420963 | |
2018 | PBMC Allogenic | K562-mbIL15-41BBL. | Relapsed or Refractory Neuroblastoma | Anti-GD2 | |
2019 | PBMC Allogenic | Recurrent Ovarian Carcinoma |
Year | Cell Source | Nanoparticle | Tumor Type | Combination | Ref. |
---|---|---|---|---|---|
2014 | Endogenous | Lipid-calcium-phosphate nanoparticle and liposome-protamine-hyaluronic acid nanoparticle | Melanoma | siTGF-β | [64] |
2012 | Endogenous | Liposomal polymeric gel | Metastatic melanoma | TGF-β inhibitor (SB505124) | [65] |
2020 | NK-92 | Nanoemulsion | Triple negative breast cancer | Selenocysteine, TGF-β inhibitor (SB505124) | [66] |
2017 | Endogenous | Chitosan nanoparticle | Colon cancer | NKG2D, IL-21 | [67] |
2018 | Endogenous | DOTAP:cholesterol nanovesicle | Lung cancer | TUSC2 gene, anti-PD-1 | [68] |
2019 | Endogenous | Lipid nanoparticle | Triple negative breast cancer | cdGMP, monophosphoryl lipid A | [69] |
2017 | Endogenous | PLGA microsphere | Hepatocellular carcinoma | IFN-γ, Transcatheter intra-arterial infusion | [70] |
2012 | NK-92MI | Magnetic nanoparticle | B cell lymphoma | External magnetic field | [71] |
2018 | Human primary NK cell | Magnetic nanoparticle | Non-small cell lung cancer | External magnetic field | [55] |
2015 | Mouse primary NK cell | TRAIL-coated liposome | Lymph node metastatic cancer | TRAIL, anti-NK1.1 | [72] |
2020 | NK-92MI | Cationic magnetic nanoparticle | Triple negative breast cancer | - | [73] |
2019 | Human primary NK cell | Immunomodulating nanoparticle | Triple negative breast cancer | phenylboronic acid, IgG | [74] |
2020 | Mouse primary NK cell | Trifunctional PLGA nanoparticle | EGFR positive solid tumor | Epirubicin | [75] |
Publisher’s Note: MDPI stays neutral with regard to jurisdictional claims in published maps and institutional affiliations. |
© 2021 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
Kim, K.-S.; Kim, D.-H.; Kim, D.-H. Recent Advances to Augment NK Cell Cancer Immunotherapy Using Nanoparticles. Pharmaceutics 2021, 13, 525. https://doi.org/10.3390/pharmaceutics13040525
Kim K-S, Kim D-H, Kim D-H. Recent Advances to Augment NK Cell Cancer Immunotherapy Using Nanoparticles. Pharmaceutics. 2021; 13(4):525. https://doi.org/10.3390/pharmaceutics13040525
Chicago/Turabian StyleKim, Kwang-Soo, Dong-Hwan Kim, and Dong-Hyun Kim. 2021. "Recent Advances to Augment NK Cell Cancer Immunotherapy Using Nanoparticles" Pharmaceutics 13, no. 4: 525. https://doi.org/10.3390/pharmaceutics13040525
APA StyleKim, K.-S., Kim, D.-H., & Kim, D.-H. (2021). Recent Advances to Augment NK Cell Cancer Immunotherapy Using Nanoparticles. Pharmaceutics, 13(4), 525. https://doi.org/10.3390/pharmaceutics13040525