Engineering the Bridge between Innate and Adaptive Immunity for Cancer Immunotherapy: Focus on γδ T and NK Cells
Abstract
:1. Introduction
2. Adoptive Cell Therapy Based on TCR Editing and CAR
2.1. TCR-Engineered T Cells
2.2. CAR Engineered T Cells
2.3. CAR Versus TCR: Pros and Cons
3. γδ T Cell Engineering Strategies
3.1. TCR Gene Transfer Involving γδ T Cells
3.2. αβ TCR Transfer into γδ T Cells
3.3. γδ TCR Transfer into αβ T Cells
3.4. iNKT TCR Transfer Into γδ T Cells
3.5. Other TCR Transfers
3.6. CAR Engineered γδ T Cells
3.6.1. Why Engineer γδ T Cells with CAR?
3.6.2. CAR γδ T Cells Engineering Strategies
3.6.3. Artificial APC/Engineered Feeder Cells
3.7. Alternative γδ T Cells Engineering Strategies
4. NK Cell Engineering Strategies
4.1. TCR-Engineered NK Cells
4.2. Source of NK Cells for CAR Development
4.3. NK Cell Transduction with CAR-Encoding Viral Vectors
5. Pre-Clinical and Clinical Applications of CAR Engineered NK Cells
5.1. Solid Tumors: Pre-Clinical Studies
5.1.1. Breast Cancers
5.1.2. Glioblastoma (GB)
5.1.3. Colorectal Cancers
5.1.4. Ewing Sarcoma
5.1.5. Hepatocellular Carcinomas
5.2. Engineered NK Cells for Hematological Malignancies
5.2.1. Leukemias and Lymphomas
- “CAR-modified pNK cells for CD7+ relapsed or refractory leukemia and lymphoma” (NCT02742727) using NK-92 cells engineered with anti-CD7 attached to TCRζ, CD28, and 4-1BB signaling domains.
- “PCAR-119 bridge immunotherapy prior to stem cell transplant in treating patients with CD19+relapsed or refractory leukemia and lymphoma” (NCT02892695) based on the use of NK-92 cell line engineered to contain anti-CD19 attached to TCRζ, CD28, and 4-1BB signaling domains.
5.2.2. Multiple Myeloma
5.2.3. Solid Tumors: Clinical Trials Using Engineered NK Cells
- “Clinical research of ROBO1 specific CAR-NK cells on patients with solid tumors” (NCT03940820). The purpose of this study is to evaluate the safety and effectiveness of cell therapy using ROBO1 CAR-NK cells in treating different types of solid tumors. The acceptance criteria for patients are the ROBO1 expression in malignancy tissues detected by immuno-histochemistry and the diagnosis of advanced solid tumor.
- “Study evaluating the efficacy and safety of CAR-modified PB NK cells in MUC1+ advanced refractory or relapsed solid tumors” (NCT02839954).
- “Clinical research of ROBO1-specific BiCAR-NK cells on patients with pancreatic cancer” (NCT03941457).
6. Conclusions
Author Contributions
Funding
Conflicts of Interest
References
- Foppen, M.G.; Donia, M.; Svane, I.M.; Haanen, J.B.A.G. Tumor-infiltrating lymphocytes for the treatment of metastatic cancer. Mol. Oncol. 2015, 9, 1918–1935. [Google Scholar] [CrossRef]
- Eberlein, T.J.; Rosenstein, M.; Rosenberg, S.A. Regression of a disseminated syngeneic solid tumor by systemic transfer of lymphoid cells expanded in interleukin 2. J. Exp. Med. 1982, 156, 385–397. [Google Scholar] [CrossRef] [Green Version]
- Stevanović, S.; Draper, L.M.; Langhan, M.M.; Campbell, T.E.; Kwong, M.L.; Wunderlich, J.R.; Dudley, M.E.; Yang, J.C.; Sherry, R.M.; Kammula, U.S.; et al. Complete regression of metastatic cervical cancer after treatment with human papillomavirus-targeted tumor-infiltrating T cells. J. Clin. Oncol. 2015, 33, 1543–1550. [Google Scholar] [CrossRef] [Green Version]
- Tran, E.; Turcotte, S.; Gros, A.; Robbins, P.F.; Lu, Y.C.; Dudley, M.E.; Wunderlich, J.R.; Somerville, R.P.; Hogan, K.; Hinrichs, C.S.; et al. Cancer immunotherapy based on mutation-specific CD4+ T cells in a patient with epithelial cancer. Science 2014, 344, 641–645. [Google Scholar] [CrossRef]
- Hopewell, E.L.; Cox, C.; Pilon-Thomas, S.; Kelley, L.L. Tumor-infiltrating lymphocytes: Streamlining a complex manufacturing process. Cytotherapy 2019, 21, 307–314. [Google Scholar] [CrossRef] [PubMed]
- Fesnak, A.D.; June, C.H.; Levine, B.L. Engineered T cells: The promise and challenges of cancer immunotherapy. Nat. Rev. Cancer 2016, 16, 566–581. [Google Scholar] [CrossRef] [PubMed]
- Caligiuri, M.A. Human natural killer cells. Blood 2008, 112, 461–469. [Google Scholar] [CrossRef] [PubMed]
- Vivier, E.; Raulet, D.H.; Moretta, A.; Caligiuri, M.A.; Zitvogel, L.; Lanier, L.L.; Yokoyama, W.M.; Ugolini, S. Innate or adaptive immunity? The example of natural killer cells. Science 2011, 331, 44–49. [Google Scholar] [CrossRef] [Green Version]
- Vivier, E.; Tomasello, E.; Baratin, M.; Walzer, T.; Ugolini, S. Functions of natural killer cells. Nat. Immunol. 2008, 9, 503–510. [Google Scholar] [CrossRef]
- Edwards, S.C.; Sutton, C.E.; Ladell, K.; Grant, E.J.; McLaren, J.E.; Roche, F.; Dash, P.; Apiwattanakul, N.; Awad, W.; Miners, K.L.; et al. A population of proinflammatory T cells coexpresses αβ and γδ T cell receptors in mice and humans. J. Exp. Med. 2020, 217. [Google Scholar] [CrossRef] [Green Version]
- Di Lorenzo, B.; Simões, A.E.; Caiado, F.; Tieppo, P.; Correia, D.V.; Carvalho, T.; da Silva, M.G.; Déchanet-Merville, J.; Schumacher, T.N.; Prinz, I.; et al. Broad Cytotoxic Targeting of Acute Myeloid Leukemia by Polyclonal Delta One T Cells. Cancer Immunol. Res. 2019, 7, 552–558. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Nussbaumer, O.; Koslowski, M. The emerging role of γδ T cells in cancer immunotherapy. Immuno Oncol. Technol. 2019, 1, 3–10. [Google Scholar] [CrossRef] [Green Version]
- Capsomidis, A.; Benthall, G.; Van Acker, H.H.; Fisher, J.; Kramer, A.M.; Abeln, Z.; Majani, Y.; Gileadi, T.; Wallace, R.; Gustafsson, K.; et al. Chimeric Antigen Receptor-Engineered Human Gamma Delta T Cells: Enhanced Cytotoxicity with Retention of Cross Presentation. Mol. Ther. 2018, 26, 354–365. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Fisher, J.; Anderson, J. Engineering Approaches in Human Gamma Delta T Cells for Cancer Immunotherapy. Front. Immunol. 2018, 9, 1409. [Google Scholar] [CrossRef]
- Silva-Santos, B.; Serre, K.; Norell, H. γδ T cells in cancer. Nat. Rev. Immunol. 2015, 15, 683–691. [Google Scholar] [CrossRef]
- June, C.H.; O’Connor, R.S.; Kawalekar, O.U.; Ghassemi, S.; Milone, M.C. CAR T cell immunotherapy for human cancer. Science 2018, 359, 1361–1365. [Google Scholar] [CrossRef] [Green Version]
- Dembic, Z.; Haas, W.; Weiss, S.; McCubrey, J.; Kiefer, H.; von Boehmer, H.; Steinmetz, M. Transfer of specificity by murine alpha and beta T-cell receptor genes. Nature 1986, 320, 232–238. [Google Scholar] [CrossRef]
- Cooper, L.J.; Kalos, M.; Lewinsohn, D.A.; Riddell, S.R.; Greenberg, P.D. Transfer of specificity for human immunodeficiency virus type 1 into primary human T lymphocytes by introduction of T-cell receptor genes. J. Virol. 2000, 74, 8207–8212. [Google Scholar] [CrossRef] [Green Version]
- Clay, T.M.; Custer, M.C.; Sachs, J.; Hwu, P.; Rosenberg, S.A.; Nishimura, M.I. Efficient transfer of a tumor antigen-reactive TCR to human peripheral blood lymphocytes confers anti-tumor reactivity. J. Immunol. 1999, 163, 507–513. [Google Scholar]
- Morgan, R.A.; Dudley, M.E.; Wunderlich, J.R.; Hughes, M.S.; Yang, J.C.; Sherry, R.M.; Royal, R.E.; Topalian, S.L.; Kammula, U.S.; Restifo, N.P.; et al. Cancer Regression in Patients After Transfer of Genetically Engineered Lymphocytes. Science 2006, 314, 126–129. [Google Scholar] [CrossRef] [Green Version]
- Rapoport, A.P.; Stadtmauer, E.A.; Binder-Scholl, G.K.; Goloubeva, O.; Vogl, D.T.; Lacey, S.F.; Badros, A.Z.; Garfall, A.; Weiss, B.; Finklestein, J.; et al. NY-ESO-1-specific TCR-engineered T cells mediate sustained antigen-specific antitumor effects in myeloma. Nat. Med. 2015, 21, 914–921. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Bendle, G.M.; Linnemann, C.; Hooijkaas, A.I.; Bies, L.; de Witte, M.A.; Jorritsma, A.; Kaiser, A.D.; Pouw, N.; Debets, R.; Kieback, E.; et al. Lethal graft-versus-host disease in mouse models of T cell receptor gene therapy. Nat. Med. 2010, 16, 565–570. [Google Scholar] [CrossRef] [PubMed]
- Van Loenen, M.M.; de Boer, R.; Amir, A.L.; Hagedoorn, R.S.; Volbeda, G.L.; Willemze, R.; van Rood, J.J.; Falkenburg, J.H.F.; Heemskerk, M.H.M. Mixed T cell receptor dimers harbor potentially harmful neoreactivity. Proc. Natl. Acad. Sci. USA 2010, 107, 10972–10977. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Liang, X.; Weigand, L.U.; Schuster, I.G.; Eppinger, E.; van der Griendt, J.C.; Schub, A.; Leisegang, M.; Sommermeyer, D.; Anderl, F.; Han, Y.; et al. A single TCR alpha-chain with dominant peptide recognition in the allorestricted HER2/neu-specific T cell repertoire. J. Immunol. 2010, 184, 1617–1629. [Google Scholar] [CrossRef] [PubMed]
- Reuß, S.; Sebestyén, Z.; Heinz, N.; Loew, R.; Baum, C.; Debets, R.; Uckert, W. TCR-engineered T cells: A model of inducible TCR expression to dissect the interrelationship between two TCRs. Eur. J. Immunol. 2014, 44, 265–274. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Cohen, C.J.; Zhao, Y.; Zheng, Z.; Rosenberg, S.A.; Morgan, R.A. Enhanced antitumor activity of murine-human hybrid T-cell receptor (TCR) in human lymphocytes is associated with improved pairing and TCR/CD3 stability. Cancer Res. 2006, 66, 8878–8886. [Google Scholar] [CrossRef] [Green Version]
- Davis, J.L.; Theoret, M.R.; Zheng, Z.; Lamers, C.H.; Rosenberg, S.A.; Morgan, R.A. Development of human anti-murine T-cell receptor antibodies in both responding and nonresponding patients enrolled in TCR gene therapy trials. Clin. Cancer Res. 2010, 16, 5852–5861. [Google Scholar] [CrossRef] [Green Version]
- Kuball, J.; Dossett, M.L.; Wolfl, M.; Ho, W.Y.; Voss, R.H.; Fowler, C.; Greenberg, P.D. Facilitating matched pairing and expression of TCR chains introduced into human T cells. Blood 2007, 109, 2331–2338. [Google Scholar] [CrossRef] [Green Version]
- Govers, C.; Sebestyén, Z.; Coccoris, M.; Willemsen, R.A.; Debets, R. T cell receptor gene therapy: Strategies for optimizing transgenic TCR pairing. Trends Mol. Med. 2010, 16, 77–87. [Google Scholar] [CrossRef]
- Albers, J.J.; Ammon, T.; Gosmann, D.; Audehm, S.; Thoene, S.; Winter, C.; Secci, R.; Wolf, A.; Stelzl, A.; Steiger, K.; et al. Gene editing enables T-cell engineering to redirect antigen specificity for potent tumor rejection. Life Sci. Alliance 2019, 2, e201900367. [Google Scholar] [CrossRef]
- Eyquem, J.; Mansilla-Soto, J.; Giavridis, T.; van der Stegen, S.J.C.; Hamieh, M.; Cunanan, K.M.; Odak, A.; Gönen, M.; Sadelain, M. Targeting a CAR to the TRAC locus with CRISPR/Cas9 enhances tumour rejection. Nature 2017, 543, 113–117. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Roth, T.L.; Puig-Saus, C.; Yu, R.; Shifrut, E.; Carnevale, J.; Li, P.J.; Hiatt, J.; Saco, J.; Krystofinski, P.; Li, H.; et al. Reprogramming human T cell function and specificity with non-viral genome targeting. Nature 2018, 559, 405–409. [Google Scholar] [CrossRef] [PubMed]
- Legut, M.; Dolton, G.; Mian, A.A.; Ottmann, O.G.; Sewell, A.K. CRISPR-mediated TCR replacement generates superior anticancer transgenic T cells. Blood 2018, 131, 311–322. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- 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, eaba7365. [Google Scholar] [CrossRef] [PubMed]
- Provasi, E.; Genovese, P.; Lombardo, A.; Magnani, Z.; Liu, P.Q.; Reik, A.; Chu, V.; Paschon, D.E.; Zhang, L.; Kuball, J.; et al. Editing T cell specificity towards leukemia by zinc finger nucleases and lentiviral gene transfer. Nat. Med. 2012, 18, 807–815. [Google Scholar] [CrossRef] [PubMed]
- Torikai, H.; Reik, A.; Liu, P.Q.; Zhou, Y.; Zhang, L.; Maiti, S.; Huls, H.; Miller, J.C.; Kebriaei, P.; Rabinovich, B.; et al. A foundation for universal T-cell based immunotherapy: T cells engineered to express a CD19-specific chimeric-antigen-receptor and eliminate expression of endogenous TCR. Blood 2012, 119, 5697–5705. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Kuwana, Y.; Asakura, Y.; Utsunomiya, N.; Nakanishi, M.; Arata, Y.; Itoh, S.; Nagase, F.; Kurosawa, Y. Expression of chimeric receptor composed of immunoglobulin-derived V regions and T-cell receptor-derived C regions. Biochem. Biophys. Res. Commun. 1987, 149, 960–968. [Google Scholar] [CrossRef]
- Gross, G.; Gorochov, G.; Waks, T.; Eshhar, Z. Generation of effector T cells expressing chimeric T cell receptor with antibody type-specificity. Transplant. Proc. 1989, 21, 127–130. [Google Scholar]
- Hartmann, J.; Schüßler-Lenz, M.; Bondanza, A.; Buchholz, C.J. Clinical development of CAR T cells-challenges and opportunities in translating innovative treatment concepts. EMBO Mol. Med. 2017, 9, 1183–1197. [Google Scholar] [CrossRef]
- Yazdanifar, M.; Zhou, R.; Mukherjee, P. Emerging immunotherapeutics in adenocarcinomas: A focus on CAR-T cells. Curr. Trends Immunol. 2016, 17, 95–115. [Google Scholar]
- Yazdanifar, M.; Zhou, R.; Grover, P.; Williams, C.; Bose, M.; Moore, L.J.; Wu, S.T.; Maher, J.; Dreau, D.; Mukherjee, A.P. Overcoming Immunological Resistance Enhances the Efficacy of A Novel Anti-tMUC1-CAR T Cell Treatment against Pancreatic Ductal Adenocarcinoma. Cells 2019, 8, 1070. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Allegra, A.; Innao, V.; Gerace, D.; Vaddinelli, D.; Musolino, C. Adoptive immunotherapy for hematological malignancies: Current status and new insights in chimeric antigen receptor T cells. Blood Cells Mol. Dis. 2016, 62, 49–63. [Google Scholar] [CrossRef] [PubMed]
- Chavez, J.C.; Bachmeier, C.; Kharfan-Dabaja, M.A. CAR T-cell therapy for B-cell lymphomas: Clinical trial results of available products. Ther. Adv. Hematol. 2019, 10, 2040620719841581. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Majzner, R.G.; Mackall, C.L. Clinical lessons learned from the first leg of the CAR T cell journey. Nat. Med. 2019, 25, 1341–1355. [Google Scholar] [CrossRef]
- Piepenbrink, K.H.; Blevins, S.J.; Scott, D.R.; Baker, B.M. The basis for limited specificity and MHC restriction in a T cell receptor interface. Nat. Commun. 2013, 4, 1–9. [Google Scholar] [CrossRef] [Green Version]
- Dotti, G.; Gottschalk, S.; Savoldo, B.; Brenner, M.K. Design and development of therapies using chimeric antigen receptor-expressing T cells. Immunol. Rev. 2014, 257, 107–126. [Google Scholar] [CrossRef]
- 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]
- Harris, D.T.; Kranz, D.M. Adoptive T Cell Therapies: A Comparison of T Cell Receptors and Chimeric Antigen Receptors. Trends Pharm. Sci. 2016, 37, 220–230. [Google Scholar] [CrossRef] [Green Version]
- Patel, K.; Olivares, S.; Singh, H.; Hurton, L.V.; Huls, M.H.; Qazilbash, M.H.; Kebriaei, P.; Champlin, R.E.; Cooper, L.J.N. Combination Immunotherapy with NY-ESO-1-Specific CAR+ T Cells with T-Cell Vaccine Improves Anti-Myeloma Effect. Blood 2016, 128, 3366. [Google Scholar] [CrossRef]
- Walseng, E.; Koksal, H.; Sektioglu, I.M.; Fane, A.; Skorstad, G.; Kvalheim, G.; Gaudernack, G.; Inderberg, E.M.; Walchli, S. A TCR-based Chimeric Antigen Receptor. Sci. Rep. 2017, 7, 10713. [Google Scholar] [CrossRef]
- Gomes, A.Q.; Correia, D.V.; Silva-Santos, B. Non-classical major histocompatibility complex proteins as determinants of tumour immunosurveillance. EMBO Rep. 2007, 8, 1024–1030. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Van der Veken, L.T.; Coccoris, M.; Swart, E.; Falkenburg, J.H.; Schumacher, T.N.; Heemskerk, M.H. Alpha beta T cell receptor transfer to gamma delta T cells generates functional effector cells without mixed TCR dimers in vivo. J. Immunol. 2009, 182, 164–170. [Google Scholar] [CrossRef] [PubMed]
- Sebestyen, Z.; Prinz, I.; Déchanet-Merville, J.; Silva-Santos, B.; Kuball, J. Translating gammadelta (γδ) T cells and their receptors into cancer cell therapies. Nat. Rev. Drug Discov. 2020, 19, 169–184. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Dorrie, J.; Krug, C.; Hofmann, C.; Muller, I.; Wellner, V.; Knippertz, I.; Schierer, S.; Thomas, S.; Zipperer, E.; Printz, D.; et al. Human adenovirus-specific gamma/delta and CD8+ T cells generated by T-cell receptor transfection to treat adenovirus infection after allogeneic stem cell transplantation. PLoS ONE 2014, 9, e109944. [Google Scholar] [CrossRef] [PubMed]
- Harrer, D.C.; Simon, B.; Fujii, S.I.; Shimizu, K.; Uslu, U.; Schuler, G.; Gerer, K.F.; Hoyer, S.; Dorrie, J.; Schaft, N. RNA-transfection of gamma/delta T cells with a chimeric antigen receptor or an alpha/beta T-cell receptor: A safer alternative to genetically engineered alpha/beta T cells for the immunotherapy of melanoma. BMC Cancer 2017, 17, 551. [Google Scholar] [CrossRef]
- Van der Veken, L.T.; Hagedoorn, R.S.; van Loenen, M.M.; Willemze, R.; Falkenburg, J.H.; Heemskerk, M.H. Alphabeta T-cell receptor engineered gammadelta T cells mediate effective antileukemic reactivity. Cancer Res. 2006, 66, 3331–3337. [Google Scholar] [CrossRef] [Green Version]
- Hiasa, A.; Nishikawa, H.; Hirayama, M.; Kitano, S.; Okamoto, S.; Chono, H.; Yu, S.S.; Mineno, J.; Tanaka, Y.; Minato, N.; et al. Rapid alphabeta TCR-mediated responses in gammadelta T cells transduced with cancer-specific TCR genes. Gene Ther. 2009, 16, 620–628. [Google Scholar] [CrossRef] [Green Version]
- Marcu-Malina, V.; Heijhuurs, S.; van Buuren, M.; Hartkamp, L.; Strand, S.; Sebestyen, Z.; Scholten, K.; Martens, A.; Kuball, J. Redirecting alphabeta T cells against cancer cells by transfer of a broadly tumor-reactive gammadeltaT-cell receptor. Blood 2011, 118, 50–59. [Google Scholar] [CrossRef]
- Allison, T.J.; Winter, C.C.; Fournie, J.J.; Bonneville, M.; Garboczi, D.N. Structure of a human gammadelta T-cell antigen receptor. Nature 2001, 411, 820–824. [Google Scholar] [CrossRef] [Green Version]
- Fisher, J.P.; Yan, M.; Heuijerjans, J.; Carter, L.; Abolhassani, A.; Frosch, J.; Wallace, R.; Flutter, B.; Capsomidis, A.; Hubank, M.; et al. Neuroblastoma killing properties of Vdelta2 and Vdelta2-negative gammadeltaT cells following expansion by artificial antigen-presenting cells. Clin. Cancer Res. 2014, 20, 5720–5732. [Google Scholar] [CrossRef] [Green Version]
- Grunder, C.; van Dorp, S.; Hol, S.; Drent, E.; Straetemans, T.; Heijhuurs, S.; Scholten, K.; Scheper, W.; Sebestyen, Z.; Martens, A.; et al. gamma9 and delta2CDR3 domains regulate functional avidity of T cells harboring gamma9delta2TCRs. Blood 2012, 120, 5153–5162. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Straetemans, T.; Grunder, C.; Heijhuurs, S.; Hol, S.; Slaper-Cortenbach, I.; Bonig, H.; Sebestyen, Z.; Kuball, J. Untouched GMP-Ready Purified Engineered Immune Cells to Treat Cancer. Clin. Cancer Res. 2015, 21, 3957–3968. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Kerzerho, J.; Yu, E.D.; Barra, C.M.; Alari-Pahissa, E.; Girardi, E.; Harrak, Y.; Lauzurica, P.; Llebaria, A.; Zajonc, D.M.; Akbari, O.; et al. Structural and functional characterization of a novel nonglycosidic type I NKT agonist with immunomodulatory properties. J. Immunol. 2012, 188, 2254–2265. [Google Scholar] [CrossRef] [PubMed]
- Kohlgruber, A.C.; Donado, C.A.; LaMarche, N.M.; Brenner, M.B.; Brennan, P.J. Activation strategies for invariant natural killer T cells. Immunogenetics 2016, 68, 649–663. [Google Scholar] [CrossRef]
- Takeda, K.; Hayakawa, Y.; Van Kaer, L.; Matsuda, H.; Yagita, H.; Okumura, K. Critical contribution of liver natural killer T cells to a murine model of hepatitis. Proc. Natl. Acad. Sci. USA 2000, 97, 5498–5503. [Google Scholar] [CrossRef] [Green Version]
- Qin, Y.; Oh, S.; Lim, S.; Shin, J.H.; Yoon, M.S.; Park, S.-H. Invariant NKT cells facilitate cytotoxic T-cell activation via direct recognition of CD1d on T cells. Exp. Mol. Med. 2019, 51, 1–9. [Google Scholar] [CrossRef] [Green Version]
- Fujii, S.I.; Shimizu, K. Immune Networks and Therapeutic Targeting of iNKT Cells in Cancer. Trends Immunol. 2019, 40, 984–997. [Google Scholar] [CrossRef]
- Liu, D.; Song, L.; Brawley, V.S.; Robison, N.; Wei, J.; Gao, X.; Tian, G.; Margol, A.; Ahmed, N.; Asgharzadeh, S.; et al. Medulloblastoma expresses CD1d and can be targeted for immunotherapy with NKT cells. Clin. Immunol. 2013, 149, 55–64. [Google Scholar] [CrossRef] [Green Version]
- Motohashi, S.; Okamoto, Y.; Yoshino, I.; Nakayama, T. Anti-tumor immune responses induced by iNKT cell-based immunotherapy for lung cancer and head and neck cancer. Clin. Immunol. 2011, 140, 167–176. [Google Scholar] [CrossRef]
- Tachibana, T.; Onodera, H.; Tsuruyama, T.; Mori, A.; Nagayama, S.; Hiai, H.; Imamura, M. Increased intratumor Valpha24-positive natural killer T cells: A prognostic factor for primary colorectal carcinomas. Clin. Cancer Res. 2005, 11, 7322–7327. [Google Scholar] [CrossRef] [Green Version]
- Kawano, T.; Cui, J.; Koezuka, Y.; Toura, I.; Kaneko, Y.; Motoki, K.; Ueno, H.; Nakagawa, R.; Sato, H.; Kondo, E.; et al. CD1d-restricted and TCR-mediated activation of valpha14 NKT cells by glycosylceramides. Science 1997, 278, 1626–1629. [Google Scholar] [CrossRef] [PubMed]
- Chang, D.H.; Osman, K.; Connolly, J.; Kukreja, A.; Krasovsky, J.; Pack, M.; Hutchinson, A.; Geller, M.; Liu, N.; Annable, R.; et al. Sustained expansion of NKT cells and antigen-specific T cells after injection of alpha-galactosyl-ceramide loaded mature dendritic cells in cancer patients. J. Exp. Med. 2005, 201, 1503–1517. [Google Scholar] [CrossRef] [PubMed]
- Iyoda, T.; Yamasaki, S.; Hidaka, M.; Kawano, F.; Abe, Y.; Suzuki, K.; Kadowaki, N.; Shimizu, K.; Fujii, S.I. Amelioration of NK cell function driven by Vα24(+) invariant NKT cell activation in multiple myeloma. Clin. Immunol. 2018, 187, 76–84. [Google Scholar] [CrossRef] [PubMed]
- Richter, J.; Neparidze, N.; Zhang, L.; Nair, S.; Monesmith, T.; Sundaram, R.; Miesowicz, F.; Dhodapkar, K.M.; Dhodapkar, M.V. Clinical regressions and broad immune activation following combination therapy targeting human NKT cells in myeloma. Blood 2013, 121, 423–430. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Wolf, B.J.; Choi, J.E.; Exley, M.A. Novel Approaches to Exploiting Invariant NKT Cells in Cancer Immunotherapy. Front. Immunol. 2018, 9, 384. [Google Scholar] [CrossRef] [PubMed]
- Shimizu, K.; Shinga, J.; Yamasaki, S.; Kawamura, M.; Dorrie, J.; Schaft, N.; Sato, Y.; Iyoda, T.; Fujii, S. Transfer of mRNA Encoding Invariant NKT Cell Receptors Imparts Glycolipid Specific Responses to T Cells and gammadeltaT Cells. PLoS ONE 2015, 10, e0131477. [Google Scholar] [CrossRef] [Green Version]
- Janssen, A.; Villacorta Hidalgo, J.; Beringer, D.X.; van Dooremalen, S.; Fernando, F.; van Diest, E.; Terrizi, A.R.; Bronsert, P.; Kock, S.; Schmitt-Graff, A.; et al. gammadelta T-cell Receptors Derived from Breast Cancer-Infiltrating T Lymphocytes Mediate Antitumor Reactivity. Cancer Immunol. Res. 2020, 8, 530–543. [Google Scholar] [CrossRef]
- Brandes, M.; Willimann, K.; Bioley, G.; Lévy, N.; Eberl, M.; Luo, M.; Tampé, R.; Lévy, F.; Romero, P.; Moser, B. Cross-presenting human gammadelta T cells induce robust CD8+ alphabeta T cell responses. Proc. Natl. Acad. Sci. USA 2009, 106, 2307–2312. [Google Scholar] [CrossRef] [Green Version]
- Khan, M.W.A.; Curbishley, S.M.; Chen, H.C.; Thomas, A.D.; Pircher, H.; Mavilio, D.; Steven, N.M.; Eberl, M.; Moser, B. Expanded Human Blood-Derived γδT Cells Display Potent Antigen-Presentation Functions. Front. Immunol. 2014, 5, 344. [Google Scholar] [CrossRef] [Green Version]
- Bertaina, A.; Zorzoli, A.; Petretto, A.; Barbarito, G.; Inglese, E.; Merli, P.; Lavarello, C.; Brescia, L.P.; De Angelis, B.; Tripodi, G.; et al. Zoledronic acid boosts γδ T-cell activity in children receiving αβ+ T and CD19+ cell-depleted grafts from an HLA-haplo-identical donor. Oncoimmunology 2017, 6, e1216291. [Google Scholar] [CrossRef] [Green Version]
- Kobayashi, H.; Tanaka, Y.; Yagi, J.; Minato, N.; Tanabe, K. Phase I/II study of adoptive transfer of γδ T cells in combination with zoledronic acid and IL-2 to patients with advanced renal cell carcinoma. Cancer Immunol. Immunother 2011, 60, 1075–1084. [Google Scholar] [CrossRef] [PubMed]
- Pauza, C.D.; Liou, M.-L.; Lahusen, T.; Xiao, L.; Lapidus, R.G.; Cairo, C.; Li, H. Gamma Delta T Cell Therapy for Cancer: It Is Good to be Local. Front. Immunol. 2018, 9. [Google Scholar] [CrossRef] [PubMed]
- Wada, I.; Matsushita, H.; Noji, S.; Mori, K.; Yamashita, H.; Nomura, S.; Shimizu, N.; Seto, Y.; Kakimi, K. Intraperitoneal injection of in vitro expanded Vγ9Vδ2 T cells together with zoledronate for the treatment of malignant ascites due to gastric cancer. Cancer Med. 2014, 3, 362–375. [Google Scholar] [CrossRef] [PubMed]
- Wilhelm, M.; Kunzmann, V.; Eckstein, S.; Reimer, P.; Weissinger, F.; Ruediger, T.; Tony, H.P. Gammadelta T cells for immune therapy of patients with lymphoid malignancies. Blood 2003, 102, 200–206. [Google Scholar] [CrossRef] [Green Version]
- DeBarros, A.; Chaves-Ferreira, M.; d’Orey, F.; Ribot, J.C.; Silva-Santos, B. CD70-CD27 interactions provide survival and proliferative signals that regulate T cell receptor-driven activation of human gammadelta peripheral blood lymphocytes. Eur. J. Immunol. 2011, 41, 195–201. [Google Scholar] [CrossRef]
- Ribot, J.C.; Debarros, A.; Mancio-Silva, L.; Pamplona, A.; Silva-Santos, B. B7-CD28 costimulatory signals control the survival and proliferation of murine and human gammadelta T cells via IL-2 production. J. Immunol. 2012, 189, 1202–1208. [Google Scholar] [CrossRef] [Green Version]
- Maniar, A.; Zhang, X.; Lin, W.; Gastman, B.R.; Pauza, C.D.; Strome, S.E.; Chapoval, A.I. Human gammadelta T lymphocytes induce robust NK cell-mediated antitumor cytotoxicity through CD137 engagement. Blood 2010, 116, 1726–1733. [Google Scholar] [CrossRef]
- Raffaghello, L.; Prigione, I.; Airoldi, I.; Camoriano, M.; Morandi, F.; Bocca, P.; Gambini, C.; Ferrone, S.; Pistoia, V. Mechanisms of immune evasion of human neuroblastoma. Cancer Lett. 2005, 228, 155–161. [Google Scholar] [CrossRef]
- Fisher, J.; Abramowski, P.; Wisidagamage Don, N.D.; Flutter, B.; Capsomidis, A.; Cheung, G.W.; Gustafsson, K.; Anderson, J. Avoidance of On-Target Off-Tumor Activation Using a Co-stimulation-Only Chimeric Antigen Receptor. Mol. Ther. 2017, 25, 1234–1247. [Google Scholar] [CrossRef] [Green Version]
- Fisher, J.P.; Flutter, B.; Wesemann, F.; Frosch, J.; Rossig, C.; Gustafsson, K.; Anderson, J. Effective combination treatment of GD2-expressing neuroblastoma and Ewing’s sarcoma using anti-GD2 ch14.18/CHO antibody with Vgamma9Vdelta2+ gammadeltaT cells. Oncoimmunology 2016, 5, e1025194. [Google Scholar] [CrossRef] [Green Version]
- Meuter, S.; Eberl, M.; Moser, B. Prolonged antigen survival and cytosolic export in cross-presenting human γδ T cells. Proc. Natl. Acad. Sci. USA 2010, 107, 8730–8735. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Himoudi, N.; Morgenstern, D.A.; Yan, M.; Vernay, B.; Saraiva, L.; Wu, Y.; Cohen, C.J.; Gustafsson, K.; Anderson, J. Human gammadelta T lymphocytes are licensed for professional antigen presentation by interaction with opsonized target cells. J. Immunol. 2012, 188, 1708–1716. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Brandes, M.; Willimann, K.; Moser, B. Professional antigen-presentation function by human gammadelta T Cells. Science 2005, 309, 264–268. [Google Scholar] [CrossRef]
- Anderson, J.; Gustafsson, K.; Himoudi, N.; Yan, M.; Heuijerjans, J. Licensing of gammadeltaT cells for professional antigen presentation: A new role for antibodies in regulation of antitumor immune responses. Oncoimmunology 2012, 1, 1652–1654. [Google Scholar] [CrossRef] [Green Version]
- Rischer, M.; Pscherer, S.; Duwe, S.; Vormoor, J.; Jurgens, H.; Rossig, C. Human gammadelta T cells as mediators of chimaeric-receptor redirected anti-tumour immunity. Br. J. Haematol. 2004, 126, 583–592. [Google Scholar] [CrossRef] [PubMed]
- L’Faqihi, F.E.; Guiraud, M.; Dastugue, N.; Brousset, P.; Le Bouteiller, P.; Halary, F.; Bonneville, M.; Fournie, J.J.; Champagne, E. Acquisition of a stimulatory activity for Vgamma9/Vdelta2 T cells by a Burkitt’s lymphoma cell line without loss of HLA class I expression. Hum. Immunol. 1999, 60, 928–938. [Google Scholar] [CrossRef]
- Green, A.E.; Lissina, A.; Hutchinson, S.L.; Hewitt, R.E.; Temple, B.; James, D.; Boulter, J.M.; Price, D.A.; Sewell, A.K. Recognition of nonpeptide antigens by human V gamma 9V delta 2 T cells requires contact with cells of human origin. Clin. Exp. Immunol. 2004, 136, 472–482. [Google Scholar] [CrossRef]
- Kato, Y.; Tanaka, Y.; Tanaka, H.; Yamashita, S.; Minato, N. Requirement of species-specific interactions for the activation of human gamma delta T cells by pamidronate. J. Immunol. 2003, 170, 3608–3613. [Google Scholar] [CrossRef] [Green Version]
- Scotet, E.; Martinez, L.O.; Grant, E.; Barbaras, R.; Jeno, P.; Guiraud, M.; Monsarrat, B.; Saulquin, X.; Maillet, S.; Esteve, J.P.; et al. Tumor recognition following Vgamma9Vdelta2 T cell receptor interactions with a surface F1-ATPase-related structure and apolipoprotein A-I. Immunity 2005, 22, 71–80. [Google Scholar] [CrossRef] [Green Version]
- Fisher, J.; Sharma, R.; Don, D.W.; Barisa, M.; Hurtado, M.O.; Abramowski, P.; Porter, L.; Day, W.; Borea, R.; Inglott, S.; et al. Engineering gammadeltaT cells limits tonic signaling associated with chimeric antigen receptors. Sci. Signal. 2019, 12. [Google Scholar] [CrossRef]
- Lo Presti, E.; Pizzolato, G.; Corsale, A.M.; Caccamo, N.; Sireci, G.; Dieli, F.; Meraviglia, S. gammadelta T Cells and Tumor Microenvironment: From Immunosurveillance to Tumor Evasion. Front. Immunol. 2018, 9, 1395. [Google Scholar] [CrossRef] [PubMed]
- Mirzaei, H.R.; Mirzaei, H.; Lee, S.Y.; Hadjati, J.; Till, B.G. Prospects for chimeric antigen receptor (CAR) gammadelta T cells: A potential game changer for adoptive T cell cancer immunotherapy. Cancer Lett. 2016, 380, 413–423. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Yazdanifar, M.; Barbarito, G.; Bertaina, A.; Airoldi, I. γδ T Cells: The Ideal Tool for Cancer Immunotherapy. Cells 2020, 9, 1305. [Google Scholar] [CrossRef] [PubMed]
- Suhoski, M.M.; Golovina, T.N.; Aqui, N.A.; Tai, V.C.; Varela-Rohena, A.; Milone, M.C.; Carroll, R.G.; Riley, J.L.; June, C.H. Engineering artificial antigen-presenting cells to express a diverse array of co-stimulatory molecules. Mol. Ther. 2007, 15, 981–988. [Google Scholar] [CrossRef] [PubMed]
- Deniger, D.C.; Maiti, S.N.; Mi, T.; Switzer, K.C.; Ramachandran, V.; Hurton, L.V.; Ang, S.; Olivares, S.; Rabinovich, B.A.; Huls, M.H.; et al. Activating and propagating polyclonal gamma delta T cells with broad specificity for malignancies. Clin. Cancer Res. 2014, 20, 5708–5719. [Google Scholar] [CrossRef] [Green Version]
- Cho, H.W.; Kim, S.Y.; Sohn, D.H.; Lee, M.J.; Park, M.Y.; Sohn, H.J.; Cho, H.I.; Kim, T.G. Triple costimulation via CD80, 4-1BB, and CD83 ligand elicits the long-term growth of Vgamma9Vdelta2 T cells in low levels of IL-2. J. Leukoc. Biol. 2016, 99, 521–529. [Google Scholar] [CrossRef] [Green Version]
- Wang, X.; Liu, J.; Gao, H.; Mo, X.D.; Han, T.; Xu, L.P.; Zhang, X.H.; Huang, X.J. Dendritic Cells Are Critical for the Activation and Expansion of Vdelta2(+) T Cells After Allogeneic Hematopoietic Transplantation. Front. Immunol. 2018, 9, 2528. [Google Scholar] [CrossRef] [Green Version]
- Polito, V.A.; Cristantielli, R.; Weber, G.; Del Bufalo, F.; Belardinilli, T.; Arnone, C.M.; Petretto, A.; Antonucci, L.; Giorda, E.; Tumino, N.; et al. Universal Ready-to-Use Immunotherapeutic Approach for the Treatment of Cancer: Expanded and Activated Polyclonal gammadelta Memory T Cells. Front. Immunol. 2019, 10, 2717. [Google Scholar] [CrossRef] [Green Version]
- Lapteva, N.; Durett, A.G.; Sun, J.L.; Rollins, L.A.; Huye, L.L.; Fang, J.; Dandekar, V.; Mei, Z.Y.; Jackson, K.; Vera, J.; et al. Large-scale ex vivo expansion and characterization of natural killer cells for clinical applications. Cytotherapy 2012, 14, 1131–1143. [Google Scholar] [CrossRef] [Green Version]
- Xiao, L.; Chen, C.; Li, Z.D.; Zhu, S.M.; Tay, J.C.K.; Zhang, X.; Zha, S.J.; Zeng, J.M.; Tan, W.K.; Liu, X.; et al. Large-scale expansion of V gamma 9V delta 2 T cells with engineered K562 feeder cells in G-Rex vessels and their use as chimeric antigen receptor-modified effector cells. Cytotherapy 2018, 20, 420–435. [Google Scholar] [CrossRef]
- Lamb, L.S.; Bowersock, J.; Dasgupta, A.; Gillespie, G.Y.; Su, Y.; Johnson, A.; Spencer, H.T. Engineered Drug Resistant gamma delta T Cells Kill Glioblastoma Cell Lines during a Chemotherapy Challenge: A Strategy for Combining Chemo- and Immunotherapy. PLoS ONE 2013, 8, e51805. [Google Scholar] [CrossRef]
- Sockolosky, J.T.; Trotta, E.; Parisi, G.; Picton, L.; Su, L.L.; Le, A.C.; Chhabra, A.; Silveria, S.L.; George, B.M.; King, I.C.; et al. Selective targeting of engineered T cells using orthogonal IL-2 cytokine-receptor complexes. Science 2018, 359, 1037–1042. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Cieri, N.; Camisa, B.; Cocchiarella, F.; Forcato, M.; Oliveira, G.; Provasi, E.; Bondanza, A.; Bordignon, C.; Peccatori, J.; Ciceri, F.; et al. IL-7 and IL-15 instruct the generation of human memory stem T cells from naive precursors. Blood 2013, 121, 573–584. [Google Scholar] [CrossRef] [PubMed]
- Rezvani, K.; Rouce, R.; Liu, E.; Shpall, E. Engineering Natural Killer Cells for Cancer Immunotherapy. Mol. Ther. J. Am. Soc. Gene Ther. 2017, 25, 1769–1781. [Google Scholar] [CrossRef] [PubMed]
- Zeng, J.; Tang, S.Y.; Toh, L.L.; Wang, S. Generation of “Off-the-Shelf” Natural Killer Cells from Peripheral Blood Cell-Derived Induced Pluripotent Stem Cells. Stem Cell Rep. 2017, 9, 1796–1812. [Google Scholar] [CrossRef] [Green Version]
- Rezvani, K.; Rouce, R.H. The Application of Natural Killer Cell Immunotherapy for the Treatment of Cancer. Front. Immunol. 2015, 6, 578. [Google Scholar] [CrossRef] [Green Version]
- Mensali, N.; Dillard, P.; Hebeisen, M.; Lorenz, S.; Theodossiou, T.; Myhre, M.R.; Fane, A.; Gaudernack, G.; Kvalheim, G.; Myklebust, J.H.; et al. NK cells specifically TCR-dressed to kill cancer cells. EBioMedicine 2019, 40, 106–117. [Google Scholar] [CrossRef] [Green Version]
- Parlar, A.; Sayitoglu, E.C.; Ozkazanc, D.; Georgoudaki, A.M.; Pamukcu, C.; Aras, M.; Josey, B.J.; Chrobok, M.; Branecki, S.; Zahedimaram, P.; et al. Engineering antigen-specific NK cell lines against the melanoma-associated antigen tyrosinase via TCR gene transfer. Eur. J. Immunol. 2019, 49, 1278–1290. [Google Scholar] [CrossRef]
- Knorr, D.A.; Ni, Z.; Hermanson, D.; Hexum, M.K.; Bendzick, L.; Cooper, L.J.; Lee, D.A.; Kaufman, D.S. Clinical-scale derivation of natural killer cells from human pluripotent stem cells for cancer therapy. Stem Cells Transl. Med. 2013, 2, 274–283. [Google Scholar] [CrossRef]
- Wilber, A.; Linehan, J.L.; Tian, X.; Woll, P.S.; Morris, J.K.; Belur, L.R.; McIvor, R.S.; Kaufman, D.S. Efficient and stable transgene expression in human embryonic stem cells using transposon-mediated gene transfer. Stem Cells 2007, 25, 2919–2927. [Google Scholar] [CrossRef]
- Matsuo, Y.; Drexler, H.G. Immunoprofiling of cell lines derived from natural killer-cell and natural killer-like T-cell leukemia-lymphoma. Leuk Res. 2003, 27, 935–945. [Google Scholar] [CrossRef]
- 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] [PubMed] [Green Version]
- Miller, J.P.; Perry, E.H.; Price, T.H.; Bolan, C.D., Jr.; Karanes, C.; Boyd, T.M.; Chitphakdithai, P.; King, R.J. Recovery and safety profiles of marrow and PBSC donors: Experience of the National Marrow Donor Program. Biol. Blood Marrow. Transplant. 2008, 14, 29–36. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Yuan, S.; Ziman, A.; Smeltzer, B.; Lu, Q.; Goldfinger, D. Moderate and severe adverse events associated with apheresis donations: Incidences and risk factors. Transfusion 2010, 50, 478–486. [Google Scholar] [CrossRef] [PubMed]
- Oelsner, S.; Friede, M.E.; Zhang, C.; Wagner, J.; Badura, S.; Bader, P.; Ullrich, E.; Ottmann, O.G.; Klingemann, H.; Tonn, T.; et al. Continuously expanding CAR NK-92 cells display selective cytotoxicity against B-cell leukemia and lymphoma. Cytotherapy 2017, 19, 235–249. [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 2016, 5, e1119354. [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]
- Romanski, A.; Uherek, C.; Bug, G.; Seifried, E.; Klingemann, H.; Wels, W.S.; Ottmann, O.G.; Tonn, T. CD19-CAR engineered NK-92 cells are sufficient to overcome NK cell resistance in B-cell malignancies. J. Cell Mol. Med. 2016, 20, 1287–1294. [Google Scholar] [CrossRef]
- Zhang, C.; Burger, M.C.; Jennewein, L.; Genssler, S.; Schonfeld, 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]
- 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. 2016, 65, 485–492. [Google Scholar] [CrossRef] [PubMed]
- Kobayashi, E.; Kishi, H.; Ozawa, T.; Hamana, H.; Nakagawa, H.; Jin, A.; Lin, Z.; Muraguchi, A. A chimeric antigen receptor for TRAIL-receptor 1 induces apoptosis in various types of tumor cells. Biochem. Biophys. Res. Commun. 2014, 453, 798–803. [Google Scholar] [CrossRef] [PubMed]
- 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] [PubMed]
- Gong, J.H.; Maki, G.; Klingemann, H.G. Characterization of a human cell line (NK-92) with phenotypical and functional characteristics of activated natural killer cells. Leukemia 1994, 8, 652–658. [Google Scholar] [PubMed]
- Tonn, T.; Becker, S.; Esser, R.; Schwabe, D.; Seifried, E. Cellular immunotherapy of malignancies using the clonal natural killer cell line NK-92. J. Hematother. Stem Cell Res. 2001, 10, 535–544. [Google Scholar] [CrossRef]
- Arai, S.; Meagher, R.; Swearingen, M.; Myint, H.; Rich, E.; Martinson, J.; Klingemann, H. Infusion of the allogeneic cell line NK-92 in patients with advanced renal cell cancer or melanoma: A phase I trial. Cytotherapy 2008, 10, 625–632. [Google Scholar] [CrossRef]
- Boyiadzis, M.; Agha, M.; Redner, R.L.; Sehgal, A.; Im, A.; Hou, J.Z.; Farah, R.; Dorritie, K.A.; Raptis, A.; Lim, S.H.; et al. Phase 1 clinical trial of adoptive immunotherapy using “off-the-shelf” activated natural killer cells in patients with refractory and relapsed acute myeloid leukemia. Cytotherapy 2017, 19, 1225–1232. [Google Scholar] [CrossRef]
- Tonn, T.; Schwabe, D.; Klingemann, H.G.; Becker, S.; Esser, R.; Koehl, U.; Suttorp, M.; Seifried, E.; Ottmann, O.G.; Bug, G. Treatment of patients with advanced cancer with the natural killer cell line NK-92. Cytotherapy 2013, 15, 1563–1570. [Google Scholar] [CrossRef]
- Hambach, J.; Riecken, K.; Cichutek, S.; Schutze, K.; Albrecht, B.; Petry, K.; Rockendorf, J.L.; Baum, N.; Kroger, N.; Hansen, T.; et al. Targeting CD38-Expressing Multiple Myeloma and Burkitt Lymphoma Cells In Vitro with Nanobody-Based Chimeric Antigen Receptors (Nb-CARs). Cells 2020, 9, 321. [Google Scholar] [CrossRef] [Green Version]
- Esser, R.; Müller, T.; Stefes, D.; Kloess, S.; Seidel, D.; Gillies, S.D.; Aperlo-Iffland, C.; Huston, J.S.; Uherek, C.; Schönfeld, K.; et al. NK cells engineered to express a GD2 -specific antigen receptor display built-in ADCC-like activity against tumour cells of neuroectodermal origin. J. Cell Mol. Med. 2012, 16, 569–581. [Google Scholar] [CrossRef]
- Boissel, L.; Betancur-Boissel, M.; Lu, W.; Krause, D.S.; Van Etten, R.A.; Wels, W.S.; Klingemann, H. Retargeting NK-92 cells by means of CD19- and CD20-specific chimeric antigen receptors compares favorably with antibody-dependent cellular cytotoxicity. Oncoimmunology 2013, 2, e26527. [Google Scholar] [CrossRef] [Green Version]
- Zhang, Q.; Zhang, H.; Ding, J.; Liu, H.; Li, H. Combination Therapy with EpCAM-CAR-NK-92 Cells and Regorafenib against Human Colorectal Cancer Models. J. Immunol. Res. 2018, 2018, 4263520. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Yu, M.; Luo, H.; Fan, M.; Wu, X.; Shi, B.; Di, S.; Liu, Y.; Pan, Z.; Jiang, H.; Li, Z. Development of GPC3-Specific Chimeric Antigen Receptor-Engineered Natural Killer Cells for the Treatment of Hepatocellular Carcinoma. Mol. Ther. 2018, 26, 366–378. [Google Scholar] [CrossRef] [Green Version]
- Chu, J.; Deng, Y.; Benson, D.M.; He, S.; Hughes, T.; Zhang, J.; Peng, Y.; Mao, H.; Yi, L.; Ghoshal, K.; et al. CS1-specific chimeric antigen receptor (CAR)-engineered natural killer cells enhance in vitro and in vivo antitumor activity against human multiple myeloma. Leukemia 2014, 28, 917–927. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Pinz, K.G.; Yakaboski, E.; Jares, A.; Liu, H.; Firor, A.E.; Chen, K.H.; Wada, M.; Salman, H.; Tse, W.; Hagag, N.; et al. Targeting T-cell malignancies using anti-CD4 CAR NK-92 cells. Oncotarget 2017, 8, 112783–112796. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Hu, Z. Tissue factor as a new target for CAR-NK cell immunotherapy of triple-negative breast cancer. Sci. Rep. 2020, 10, 2815. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- 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]
- You, F.; Wang, Y.; Jiang, L.; Zhu, X.; Chen, D.; Yuan, L.; An, G.; Meng, H.; Yang, L. A novel CD7 chimeric antigen receptor-modified NK-92MI cell line targeting T-cell acute lymphoblastic leukemia. Am. J. Cancer Res. 2019, 9, 64–78. [Google Scholar]
- 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. [Google Scholar] [CrossRef]
- Maroto-Martín, E.; Encinas, J.; García-Ortiz, A.; Alonso, R.; Leivas, A.; Paciello, M.L.; Garrido, V.; Cedena, T.; Ugalde, L.; Powell, D.J.J.; et al. PS1209 NKG2D and bcma-car NK cells efficiently eliminate multiple myeloma cells. A comprehensive comparison between two clinically relevant CARS. HemaSphere 2019, 3, 550–551. [Google Scholar] [CrossRef]
- Müller, S.; Bexte, T.; Gebel, V.; Kalensee, F.; Stolzenberg, E.; Hartmann, J.; Koehl, U.; Schambach, A.; Wels, W.S.; Modlich, U.; et al. High Cytotoxic Efficiency of Lentivirally and Alpharetrovirally Engineered CD19-Specific Chimeric Antigen Receptor Natural Killer Cells Against Acute Lymphoblastic Leukemia. Front. Immunol. 2019, 10, 3123. [Google Scholar] [CrossRef]
- Muyldermans, S. Nanobodies: Natural single-domain antibodies. Annu. Rev. Biochem. 2013, 82, 775–797. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Kailayangiri, S.; Altvater, B.; Spurny, C.; Jamitzky, S.; Schelhaas, S.; Jacobs, A.H.; Wiek, C.; Roellecke, K.; Hanenberg, H.; Hartmann, W.; et al. Targeting Ewing sarcoma with activated and GD2-specific chimeric antigen receptor-engineered human NK cells induces upregulation of immune-inhibitory HLA-G. Oncoimmunology 2017, 6, e1250050. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- 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]
- Moasser, M.M. The oncogene HER2: Its signaling and transforming functions and its role in human cancer pathogenesis. Oncogene 2007, 26, 6469–6487. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Golán, I.; Rodríguez de la Fuente, L.; Costoya, J.A. NK Cell-Based Glioblastoma Immunotherapy. Cancers 2018, 10, 522. [Google Scholar] [CrossRef] [Green Version]
- Dekker, E.; Tanis, P.J.; Vleugels, J.L.A.; Kasi, P.M.; Wallace, M.B. Colorectal cancer. Lancet 2019, 394, 1467–1480. [Google Scholar] [CrossRef]
- Grünewald, T.G.; Cidre-Aranaz, F.; Surdez, D.; Tomazou, E.M.; de Álava, E.; Kovar, H.; Sorensen, P.H.; Delattre, O.; Dirksen, U. Ewing sarcoma. Nat. Rev. Dis. Primers 2018, 4, 1–22. [Google Scholar] [CrossRef]
- Curran, E.K.; Godfrey, J.; Kline, J. Mechanisms of Immune Tolerance in Leukemia and Lymphoma. Trends Immunol. 2017, 38, 513–525. [Google Scholar] [CrossRef]
- Ghione, P.; Moskowitz, A.J.; De Paola, N.E.K.; Horwitz, S.M.; Ruella, M. Novel Immunotherapies for T Cell Lymphoma and Leukemia. Curr. Hematol. Malig. Rep. 2018, 13, 494–506. [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. New Engl. J. Med. 2020, 382, 545–553. [Google Scholar] [CrossRef]
- Rajkumar, S.V. Multiple myeloma: Every year a new standard? Hematol. Oncol. 2019, 37 (Suppl. S1), 62–65. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Tamura, H.; Ishibashi, M.; Sunakawa, M.; Inokuchi, K. Immunotherapy for Multiple Myeloma. Cancers 2019, 11, 2009. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Hsi, E.D.; Steinle, R.; Balasa, B.; Szmania, S.; Draksharapu, A.; Shum, B.P.; Huseni, M.; Powers, D.; Nanisetti, A.; Zhang, Y.; et al. CS1, a potential new therapeutic antibody target for the treatment of multiple myeloma. Clin. Cancer Res. 2008, 14, 2775–2784. [Google Scholar] [CrossRef] [PubMed] [Green Version]
© 2020 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 (http://creativecommons.org/licenses/by/4.0/).
Share and Cite
Morandi, F.; Yazdanifar, M.; Cocco, C.; Bertaina, A.; Airoldi, I. Engineering the Bridge between Innate and Adaptive Immunity for Cancer Immunotherapy: Focus on γδ T and NK Cells. Cells 2020, 9, 1757. https://doi.org/10.3390/cells9081757
Morandi F, Yazdanifar M, Cocco C, Bertaina A, Airoldi I. Engineering the Bridge between Innate and Adaptive Immunity for Cancer Immunotherapy: Focus on γδ T and NK Cells. Cells. 2020; 9(8):1757. https://doi.org/10.3390/cells9081757
Chicago/Turabian StyleMorandi, Fabio, Mahboubeh Yazdanifar, Claudia Cocco, Alice Bertaina, and Irma Airoldi. 2020. "Engineering the Bridge between Innate and Adaptive Immunity for Cancer Immunotherapy: Focus on γδ T and NK Cells" Cells 9, no. 8: 1757. https://doi.org/10.3390/cells9081757
APA StyleMorandi, F., Yazdanifar, M., Cocco, C., Bertaina, A., & Airoldi, I. (2020). Engineering the Bridge between Innate and Adaptive Immunity for Cancer Immunotherapy: Focus on γδ T and NK Cells. Cells, 9(8), 1757. https://doi.org/10.3390/cells9081757