Realizing Innate Potential: CAR-NK Cell Therapies for Acute Myeloid Leukemia
Abstract
:Simple Summary
Abstract
1. Introduction
2. CAR-T Cell Therapy and AML
2.1. The Contrast to CD19 CAR-T
2.2. Target Antigens in AML
3. Natural Killer Cells and AML
3.1. NK Cell Functions
3.2. NK Cells in AML Immunoediting
3.3. NK Cells in ASCT
3.4. NK Cell Adoptive Cell Transfer for AML
4. CAR-NK: A Compelling Platform for AML Immunotherapy?
4.1. Principles of CAR-NK Therapies
Cell Source | Target | Disease | NCT Identifier | Status [Reports] | Location |
---|---|---|---|---|---|
Cord Blood | CD19 | B-ALL/CLL/NHL | NCT03056339 | Active [17] | MD Anderson Cancer Center |
Cord Blood | CD19 | B-ALL/CLL/NHL | NCT04796675 | Active | Huazhong University of Science and Technology |
Haplo NK | CD19 | Pediatric B-ALL | NCT00995137 | Completed | St Jude Children’s Research Hospital |
Haplo NK | CD19 | B-ALL | NCT01974479 | Suspended | National University Hospital Singapore |
Haplo NK | NKG2DL | AML/MDS | NCT04623944 | Active | Multiple Sites (USA) |
PBNK | NKG2DL | Solid Tumors | NCT03415100 | Active [130] | Guangzhou Medical University |
iPSC-NK | CD19 | CLL/NHL | NCT04245722 | Active [133] | University of Minnesota Masonic Cancer Center |
NK-92 | ROBO1 | Solid Tumors | NCT03940820 | Active | Suzhou Cancer Center |
NK-92 | ROBO1 | Pancreatic Cancer | NCT03941457 | Active [139] | Shanghai Ruijin Hospital |
NK-92 | BCMA | Multiple Myeloma | NCT03940833 | Active | Nanjing Medical University |
NK-92 | HER2 | Glioblastoma | NCT03383978 | Active | Johann Wolfgang Goethe University Hospital |
NK-92 | CD33 | AML | NCT02944162 | Completed [140] | Jiangsu Institute of Hematology |
4.2. AML Target Antigens and CAR-NK
4.3. Engineering Persistence: A Balance of Efficacy and Off-Tumor Effects
4.4. Next Steps: Optimizing CAR-NK Activity in AML
4.4.1. CAR-NK Cell Homing
4.4.2. Inhibitory Checkpoints
4.4.3. AML Ligand Expression and Dual Targeting
5. CAR-NK in the Context of AML Immunotherapy
6. Conclusions
Author Contributions
Funding
Conflicts of Interest
References
- Yamamoto, J.F.; Goodman, M.T. Patterns of leukemia incidence in the United States by subtype and demographic characteristics, 1997–2002. Cancer Causes Control 2008, 19, 379–390. [Google Scholar] [CrossRef]
- Arber, D.A.; Orazi, A.; Hasserjian, R.; Borowitz, M.J.; Le Beau, M.M.; Bloomfield, C.D.; Cazzola, M.; Vardiman, J.W. The 2016 revision to the World Health Organization classi fi cation of myeloid neoplasms and acute leukemia. Blood 2016, 127, 2391–2406. [Google Scholar] [CrossRef] [PubMed]
- Schuurhuis, G.J.; Heuser, M.; Freeman, S.; Béne, M.C.; Buccisano, F.; Cloos, J.; Grimwade, D.; Haferlach, T.; Hills, R.K.; Hourigan, C.S.; et al. Minimal/measurable residual disease in AML: A consensus document from the European LeukemiaNet MRD Working Party. Blood 2018, 131, 1275–1291. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Heuser, M.; Ofran, Y.; Boissel, N.; Brunet Mauri, S.; Craddock, C.; Janssen, J.; Wierzbowska, A.; Buske, C. Acute myeloid leukaemia in adult patients: ESMO Clinical Practice Guidelines for diagnosis, treatment and follow-up. Ann. Oncol. 2020, 31, 697–712. [Google Scholar] [CrossRef] [PubMed]
- Shlush, L.I.; Mitchell, A.; Heisler, L.; Abelson, S.; Ng, S.W.K.; Trotman-Grant, A.; Medeiros, J.J.F.; Rao-Bhatia, A.; Jaciw-Zurakowsky, I.; Marke, R.; et al. Tracing the origins of relapse in acute myeloid leukaemia to stem cells. Nature 2017, 547, 104–108. [Google Scholar] [CrossRef]
- Stone, R.M.; Mandrekar, S.J.; Sanford, B.L.; Laumann, K.; Geyer, S.; Bloomfield, C.D.; Thiede, C.; Prior, T.W.; Döhner, K.; Marcucci, G.; et al. Midostaurin plus Chemotherapy for Acute Myeloid Leukemia with a FLT3 Mutation. N. Engl. J. Med. 2017, 377, 454–464. [Google Scholar] [CrossRef] [PubMed]
- DiNardo, C.D.; Stein, E.M.; de Botton, S.; Roboz, G.J.; Altman, J.K.; Mims, A.S.; Swords, R.; Collins, R.H.; Mannis, G.N.; Pollyea, D.A.; et al. Durable Remissions with Ivosidenib in IDH1 -Mutated Relapsed or Refractory AML. N. Engl. J. Med. 2018, 378, 2386–2398. [Google Scholar] [CrossRef]
- Stein, E.M.; DiNardo, C.D.; Pollyea, D.A.; Fathi, A.T.; Roboz, G.J.; Altman, J.K.; Stone, R.M.; DeAngelo, D.J.; Levine, R.L.; Flinn, I.W.; et al. Enasidenib in mutant IDH2 relapsed or refractory acute myeloid leukemia. Blood 2017, 130, 722–731. [Google Scholar] [CrossRef]
- Pollyea, D.A.; Stevens, B.M.; Jones, C.L.; Winters, A.; Pei, S.; Minhajuddin, M.; D’Alessandro, A.; Culp-Hill, R.; Riemondy, K.A.; Gillen, A.E.; et al. Venetoclax with azacitidine disrupts energy metabolism and targets leukemia stem cells in patients with acute myeloid leukemia. Nat. Med. 2018, 24, 1859–1866. [Google Scholar] [CrossRef]
- Maude, S.L.; Laetsch, T.W.; Buechner, J.; Rives, S.; Boyer, M.; Bittencourt, H.; Bader, P.; Verneris, M.R.; Stefanski, H.E.; Myers, G.D.; et al. Tisagenlecleucel in Children and Young Adults with B-Cell Lymphoblastic Leukemia. N. Engl. J. Med. 2018, 378, 439–448. [Google Scholar] [CrossRef]
- Schuster, S.J.; Bishop, M.R.; Tam, C.S.; Waller, E.K.; Borchmann, P.; McGuirk, J.P.; Jäger, U.; Jaglowski, S.; Andreadis, C.; Westin, J.R.; et al. Tisagenlecleucel in Adult Relapsed or Refractory Diffuse Large B-Cell Lymphoma. N. Engl. J. Med. 2019, 380, 45–56. [Google Scholar] [CrossRef] [PubMed]
- Neelapu, S.S.; Locke, F.L.; Bartlett, N.L.; Lekakis, L.J.; Miklos, D.B.; Jacobson, C.A.; Braunschweig, I.; Oluwole, O.O.; Siddiqi, T.; Lin, Y.; et al. Axicabtagene Ciloleucel CAR T-Cell Therapy in Refractory Large B-Cell Lymphoma. N. Engl. J. Med. 2017, 377, 2531–2544. [Google Scholar] [CrossRef]
- June, C.H.; Sadelain, M. Chimeric Antigen Receptor Therapy. N. Engl. J. Med. 2018, 379, 64–73. [Google Scholar] [CrossRef]
- MacKay, M.; Afshinnekoo, E.; Rub, J.; Hassan, C.; Khunte, M.; Baskaran, N.; Owens, B.; Liu, L.; Roboz, G.J.; Guzman, M.L.; et al. The therapeutic landscape for cells engineered with chimeric antigen receptors. Nat. Biotechnol. 2020, 38, 233–244. [Google Scholar] [CrossRef] [PubMed]
- Raje, N.; Berdeja, J.; Lin, Y.; Siegel, D.; Jagannath, S.; Madduri, D.; Liedtke, M.; Rosenblatt, J.; Maus, M.V.; Turka, A.; et al. Anti-BCMA CAR T-Cell Therapy bb2121 in Relapsed or Refractory Multiple Myeloma. N. Engl. J. Med. 2019, 380, 1726–1737. [Google Scholar] [CrossRef]
- Cummins, K.D.; Gill, S. Chimeric antigen receptor T-cell therapy for acute myeloid leukemia: How close to reality? Haematologica 2019, 104, 1302–1308. [Google Scholar] [CrossRef] [Green Version]
- Liu, E.; Marin, D.; Banerjee, P.; MacApinlac, H.A.; Thompson, P.; Basar, R.; Kerbauy, L.N.; 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]
- 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] [PubMed] [Green Version]
- 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] [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]
- Lowdell, M.W.; Craston, R.; Samuel, D.; Wood, M.E.; O’Neill, E.; Saha, V.; Prentice, H.G. Evidence that continued remission in patients treated for acute leukaemia is dependent upon autologous natural killer cells. Br. J. Haematol. 2002, 117, 821–827. [Google Scholar] [CrossRef]
- Qasim, W.; Zhan, H.; Samarasinghe, S.; Adams, S.; Amrolia, P.; Stafford, S.; Butler, K.; Rivat, C.; Wright, G.; Somana, K.; et al. Molecular remission of infant B-ALL after infusion of universal TALEN gene-edited CAR T cells. Sci. Transl. Med. 2017, 9. [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]
- Costello, C.L.; Gregory, T.K.; Ali, S.A.; Berdeja, J.G.; Patel, K.K.; Shah, N.D.; Ostertag, E.; Martin, C.; Ghoddusi, M.; Shedlock, D.J.; et al. Phase 2 Study of the Response and Safety of P-Bcma-101 CAR-T Cells in Patients with Relapsed/Refractory (r/r) Multiple Myeloma (MM) (PRIME). Blood 2019, 134, 3184. [Google Scholar] [CrossRef]
- Kebriaei, P.; Singh, H.; Huls, M.H.; Figliola, M.J.; Bassett, R.; Olivares, S.; Jena, B.; Dawson, M.J.; Kumaresan, P.R.; Su, S.; et al. Phase i trials using sleeping beauty to generate CD19-specific CAR T cells. J. Clin. Invest. 2016, 126, 3363–3376. [Google Scholar] [CrossRef] [PubMed]
- 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]
- Li, D.; Li, X.; Zhou, W.L.; Huang, Y.; Liang, X.; Jiang, L.; Yang, X.; Sun, J.; Li, Z.; Han, W.D.; et al. Genetically engineered t cells for cancer immunotherapy. Signal Transduct. Target. Ther. 2019, 4, 1–17. [Google Scholar] [CrossRef]
- Maher, J.; Brentjens, R.J.; Gunset, G.; Rivière, I.; Sadelain, M. Human T-lymphocyte cytotoxicity and proliferation directed by a single chimeric TCRζ/CD28 receptor. Nat. Biotechnol. 2002, 20, 70–75. [Google Scholar] [CrossRef] [PubMed]
- Shah, N.N.; Fry, T.J. Mechanisms of resistance to CAR T cell therapy. Nat. Rev. Clin. Oncol. 2019, 16, 372–385. [Google Scholar] [CrossRef]
- Hu, Y.; Zhang, Y.; Zhao, H.; Wang, Y.; Nagler, A.; Chang, A.H.; Huang, H. CD19/CD22 Dual-Targeted Chimeric Antigen Receptor T-Cell Therapy for Relapsed/Refractory Aggressive B-Cell Lymphoma: A Safety and Efficacy Study. Blood 2020, 136, 34. [Google Scholar] [CrossRef]
- Abramson, J.S.; Palomba, M.L.; Gordon, L.I.; Lunning, M.A.; Wang, M.; Arnason, J.; Mehta, A.; Purev, E.; Maloney, D.G.; Andreadis, C.; et al. Lisocabtagene maraleucel for patients with relapsed or refractory large B-cell lymphomas (TRANSCEND NHL 001): A multicentre seamless design study. Lancet 2020, 396, 839–852. [Google Scholar] [CrossRef]
- Zheng, W.; O’Hear, C.E.; Alli, R.; Basham, J.H.; Abdelsamed, H.A.; Palmer, L.E.; Jones, L.L.; Youngblood, B.; Geiger, T.L. PI3K orchestration of the in vivo persistence of chimeric antigen receptor-modified T cells. Leukemia 2018, 32, 1157–1167. [Google Scholar] [CrossRef]
- Hudecek, M.; Ivics, Z. Non-viral therapeutic cell engineering with the Sleeping Beauty transposon system. Curr. Opin. Genet. Dev. 2018, 52, 100–108. [Google Scholar] [CrossRef]
- Cheever, M.A.; Allison, J.P.; Ferris, A.S.; Finn, O.J.; Hastings, B.M.; Hecht, T.T.; Mellman, I.; Prindiville, S.A.; Viner, J.L.; Weiner, L.M.; et al. The prioritization of cancer antigens: A National Cancer Institute pilot project for the acceleration of translational research. Clin. Cancer Res. 2009, 15, 5323–5337. [Google Scholar] [CrossRef] [Green Version]
- Haubner, S.; Perna, F.; Köhnke, T.; Schmidt, C.; Berman, S.; Augsberger, C.; Schnorfeil, F.M.; Krupka, C.; Lichtenegger, F.S.; Liu, X.; et al. Coexpression profile of leukemic stem cell markers for combinatorial targeted therapy in AML. Leukemia 2019, 33, 64–74. [Google Scholar] [CrossRef] [PubMed]
- Perna, F.; Berman, S.H.; Soni, R.K.; Mansilla-Soto, J.; Eyquem, J.; Hamieh, M.; Hendrickson, R.C.; Brennan, C.W.; Sadelain, M. Integrating Proteomics and Transcriptomics for Systematic Combinatorial Chimeric Antigen Receptor Therapy of AML. Cancer Cell 2017, 32, 506–519.e5. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Liu, F.; Cao, Y.; Pinz, K.; Ma, Y.; Wada, M.; Chen, K.; Ma, G.; Shen, J.; Tse, C.O.; Su, Y.; et al. First-in-Human CLL1-CD33 Compound CAR T Cell Therapy Induces Complete Remission in Patients with Refractory Acute Myeloid Leukemia: Update on Phase 1 Clinical Trial. Blood 2018, 132, 901. [Google Scholar] [CrossRef]
- Hu, Y.; Wu, Z.; Luo, Y.; Shi, J.; Yu, J.; Pu, C.; Liang, Z.; Wei, G.; Cui, Q.; Sun, J.; et al. Potent Anti-leukemia activities of Chimeric antigen Receptor-modified T cells against CD19 in Chinese patients with relapsed/refractory acute lymphocytic leukemia. Clin. Cancer Res. 2017, 23, 3297–3306. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Herrmann, H.; Sadovnik, I.; Eisenwort, G.; Rülicke, T.; Blatt, K.; Herndlhofer, S.; Willmann, M.; Stefanzl, G.; Baumgartner, S.; Greiner, G.; et al. Delineation of target expression profiles in CD34+/CD38- and CD34+/CD38+ stem and progenitor cells in AML and CML. Blood Adv. 2020, 4, 5118–5132. [Google Scholar] [CrossRef]
- Arnone, M.; Konantz, M.; Hanns, P.; Stanger, A.M.P.; Bertels, S.; Godavarthy, P.S.; Christopeit, M.; Lengerke, C. Acute myeloid leukemia stem cells: The challenges of phenotypic heterogeneity. Cancers 2020, 12, 3742. [Google Scholar] [CrossRef]
- Taussig, D.C.; Pearce, D.J.; Simpson, C.; Rohatiner, A.Z.; Lister, T.A.; Kelly, G.; Luongo, J.L.; Danet-Desnoyers, G.A.H.; Bonnet, D. Hematopoietic stem cells express multiple myeloid markers: Implications for the origin and targeted therapy of acute myeloid leukemia. Blood 2005, 106, 4086–4092. [Google Scholar] [CrossRef] [Green Version]
- Olombel, G.; Guerin, E.; Guy, J.; Perrot, J.Y.; Dumezy, F.; de Labarthe, A.; Bastie, J.N.; Legrand, O.; Raffoux, E.; Plesa, A.; et al. The level of blast CD33 expression positively impacts the effect of gemtuzumab ozogamicin in patients with acute myeloid leukemia. Blood 2016, 127, 2157–2160. [Google Scholar] [CrossRef] [PubMed]
- Stein, E.M.; Walter, R.B.; Erba, H.P.; Fathi, A.T.; Advani, A.S.; Lancet, J.E.; Ravandi, F.; Kovacsovics, T.; DeAngelo, D.J.; Bixby, D.; et al. A phase 1 trial of vadastuximab talirine as monotherapy in patients with CD33-positive acute myeloid leukemia. Blood 2018, 131, 387–396. [Google Scholar] [CrossRef] [Green Version]
- Baron, J.; Wang, E.S. Gemtuzumab ozogamicin for the treatment of acute myeloid leukemia. Expert Rev. Clin. Pharmacol. 2018, 11, 549–559. [Google Scholar] [CrossRef]
- Kenderian, S.; Ruella, M.; Shestova, O.; Klichinsky, M.; Aikawa, V.; Morrissette, J.; Scholler, J.; Song, D.; Porter, D.; Carroll, M.; et al. CD33-specific chimeric antigen receptor T cells exhibit potent preclinical activity against human acute myeloid leukemia. Leukemia 2015, 29, 1637–1647. [Google Scholar] [CrossRef] [PubMed]
- Wang, Q.S.; Wang, Y.; Lv, H.Y.; Han, Q.W.; Fan, H.; Guo, B.; Wang, L.L.; Han, W.D. Treatment of CD33-directed chimeric antigen receptor-modified T cells in one patient with relapsed and refractory acute myeloid leukemia. Mol. Ther. 2015, 23, 184–191. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Gill, S.; Tasian, S.K.; Ruella, M.; Shestova, O.; Li, Y.; Porter, D.L.; Carroll, M.; Danet-Desnoyers, G.; Scholler, J.; Grupp, S.A.; et al. Preclinical targeting of human acute myeloid leukemia and myeloablation using chimeric antigen receptor-modified T cells. Blood 2014, 123, 2343–2354. [Google Scholar] [CrossRef] [Green Version]
- Jordan, C.T.; Upchurch, D.; Szilvassy, S.J.; Guzman, M.L.; Howard, D.S.; Pettigrew, A.L.; Meyerrose, T.; Rossi, R.; Grimes, B.; Rizzieri, D.A.; et al. The interleukin-3 receptor alpha chain is a unique marker for human acute myelogenous leukemia stems cells. Leukemia 2000, 14, 1777–1784. [Google Scholar] [CrossRef] [Green Version]
- Budde, L.E.; Song, J.; Del Real, M.; Kim, Y.; Toribio, C.; Wood, B.; Wagner, J.; Marcucci, E.; Stein, A.; Marcucci, G.; et al. Abstract PR14: CD123CAR displays clinical activity in relapsed/refractory (r/r) acute myeloid leukemia (AML) and blastic plasmacytoid dendritic cell neoplasm (BPDCN): Safety and efficacy results from a phase 1 study. Cancer Immunol. Res. 2020, 8, PR14. [Google Scholar] [CrossRef]
- Budde, L.E. Chimeric antigen receptor T-cell therapy for acute myeloid leukemia: Targeting CD123. In Proceedings of the 1st European CAR T Cell Meeting, Paris, France, 14–16 February 2019. [Google Scholar]
- Kenderian, S.S.; Ruella, M.; Shestova, O.; Klichinsky, M.; Kim, M.; Soderquist, C.; Bagg, A.; Singh, R.; Richardson, C.; Young, R.; et al. Targeting CLEC12A with Chimeric Antigen Receptor T Cells Can Overcome the Chemotherapy Refractoriness of Leukemia Stem Cells. Biol. Blood Marrow Transplant. 2017, 23, S247–S248. [Google Scholar] [CrossRef]
- Ritchie, D.S.; Neeson, P.J.; Khot, A.; Peinert, S.; Tai, T.; Tainton, K.; Chen, K.; Shin, M.; Wall, D.M.; Hönemann, D.; et al. Persistence and efficacy of second generation CAR T Cell against the LeY Antigen in acute myeloid leukemia. Mol. Ther. 2013, 21, 2122–2129. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Gomes-Silva, D.; Atilla, E.; Atilla, P.A.; Mo, F.; Tashiro, H.; Srinivasan, M.; Lulla, P.; Rouce, R.H.; Cabral, J.M.S.; Ramos, C.A.; et al. CD7 CAR T Cells for the Therapy of Acute Myeloid Leukemia. Mol. Ther. 2019, 27, 272–280. [Google Scholar] [CrossRef]
- Cho, J.H.; Collins, J.J.; Wong, W.W. Universal Chimeric Antigen Receptors for Multiplexed and Logical Control of T Cell Responses. Cell 2018, 173, 1426–1438.e11. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Benmebarek, M.R.; Cadilha, B.L.; Herrmann, M.; Lesch, S.; Schmitt, S.; Stoiber, S.; Darwich, A.; Augsberger, C.; Brauchle, B.; Rohrbacher, L.; et al. A modular and controllable T cell therapy platform for acute myeloid leukemia. Leukemia 2021, 1–15. [Google Scholar] [CrossRef]
- Liu, F. First-in-Human CLL1-CD33 Compound Car (CCAR) T Cell Therapy in Relapsed and Refractory Acute Myeloid Leukemia. In Proceedings of the 25th EHA Annual Congress, Frankfurt, Germany, 11–21 June 2020; p. S149. [Google Scholar]
- Sallman, D.A.; Brayer, J.; Sagatys, E.M.; Lonez, C.; Breman, E.; Agaugué, S.; Verma, B.; Gilham, D.E.; Lehmann, F.F.; Davila, M.L. NKG2D-based chimeric antigen receptor therapy induced remission in a relapsed/refractory acute myeloid leukemia patient. Haematologica 2018, 103, e424–e426. [Google Scholar] [CrossRef] [Green Version]
- Sallman, D.A.; Brayer, J.B.; Poire, X.; Havelange, V.; Awada, A.; Lewalle, P.; Odunsi, K.; Wang, E.S.; Lonez, C.; Lequertier, T.; et al. Results from the Completed Dose-Escalation of the Hematological Arm of the Phase I Think Study Evaluating Multiple Infusions of NKG2D-Based CAR T-Cells As Standalone Therapy in Relapse/Refractory Acute Myeloid Leukemia and Myelodysplastic Syndrome Patients. Blood 2019, 134, 3826. [Google Scholar] [CrossRef]
- Sallman, D. Results from the Phase I Clinical Studies Evaluating Cyad-01, a First-Generation NKG2D CAR T-Cell Product in Relapsed or Refractory Acute Myeloid Leukemia and Myelodysplastic Syndrome Patients. In Proceedings of the 62nd ASH Annual meeting and exposition, San Diego, CA, USA, 5–8 December 2020. [Google Scholar]
- Xie, G.; Ivica, N.A.; Jia, B.; Li, Y.; Dong, H.; Liang, Y.; Brown, D.; Rizwan, R.; Chen, J. CAR-T cells targeting a nucleophosmin neoepitope exhibit potent specific activity in mouse models of acute myeloid leukaemia. Nat. Biomed. Eng. 2020. [Google Scholar] [CrossRef]
- Kim, M.Y.; Yu, K.R.; Kenderian, S.S.; Ruella, M.; Chen, S.; Shin, T.H.; Aljanahi, A.A.; Schreeder, D.; Klichinsky, M.; Shestova, O.; et al. Genetic Inactivation of CD33 in Hematopoietic Stem Cells to Enable CAR T Cell Immunotherapy for Acute Myeloid Leukemia. Cell 2018, 173, 1439–1453.e19. [Google Scholar] [CrossRef] [Green Version]
- Cummins, K.D.; Frey, N.; Nelson, A.M.; Schmidt, A.; Luger, S.; Isaacs, R.E.; Lacey, S.F.; Hexner, E.; Melenhorst, J.J.; June, C.H.; et al. Treating Relapsed / Refractory (RR) AML with Biodegradable Anti-CD123 CAR Modified T Cells. Blood 2017, 130, 1359. [Google Scholar] [CrossRef]
- Drent, E.; Themeli, M.; Poels, R.; de Jong-Korlaar, R.; Yuan, H.; de Bruijn, J.; Martens, A.C.M.; Zweegman, S.; van de Donk, N.W.C.J.; Groen, R.W.J.; et al. A Rational Strategy for Reducing On-Target Off-Tumor Effects of CD38-Chimeric Antigen Receptors by Affinity Optimization. Mol. Ther. 2017, 25, 1946–1958. [Google Scholar] [CrossRef]
- Naik, J.; Themeli, M.; de Jong-Korlaar, R.; Ruiter, R.W.J.; Poddighe, P.J.; Yuan, H.; de Bruijn, J.D.; Ossenkoppele, G.J.; Zweegman, S.; Smit, L.; et al. CD38 as a therapeutic target for adult acute myeloid leukemia and T-cell acute lymphoblastic leukemia. Haematologica 2019, 104, e100–e103. [Google Scholar] [CrossRef]
- Vivier, E.; Tomasello, E.; Baratin, M.; Walzer, T.; Ugolini, S. Functions of natural killer cells. Nat. Immunol. 2008, 9, 503–510. [Google Scholar] [CrossRef]
- Bryceson, Y.T.; Ljunggren, H.G.; Long, E.O. Minimal requirement for induction of natural cytotoxicity and intersection of activation signals by inhibitory receptors. Blood 2009, 114, 2657–2666. [Google Scholar] [CrossRef] [Green Version]
- Sheard, M.A.; Asgharzadeh, S.; Liu, Y.; Lin, T.Y.; Wu, H.W.; Ji, L.; Groshen, S.; Lee, D.A.; Seeger, R.C. Membrane-bound TRAIL supplements natural killer cell cytotoxicity against neuroblastoma cells. J. Immunother. 2013, 36, 319–329. [Google Scholar] [CrossRef] [Green Version]
- Gerosa, F.; Baldani-Guerra, B.; Nisii, C.; Marchesini, V.; Carra, G.; Trinchieri, G. Reciprocal activating interaction between natural killer cells and dendritic cells. J. Exp. Med. 2002, 195, 327–333. [Google Scholar] [CrossRef] [PubMed]
- Sag, D.; Ayyildiz, Z.O.; Gunalp, S.; Wingender, G. The role of trail/drs in the modulation of immune cells and responses. Cancers 2019, 11, 1469. [Google Scholar] [CrossRef] [Green Version]
- Hayakawa, Y.; Screpanti, V.; Yagita, H.; Grandien, A.; Ljunggren, H.-G.; Smyth, M.J.; Chambers, B.J. NK Cell TRAIL Eliminates Immature Dendritic Cells In Vivo and Limits Dendritic Cell Vaccination Efficacy. J. Immunol. 2004, 172, 123–129. [Google Scholar] [CrossRef] [PubMed]
- Boudreau, J.E.; Hsu, K.C. Natural Killer Cell Education and the Response to Infection and Cancer Therapy: Stay Tuned. Trends Immunol. 2018, 39, 222–239. [Google Scholar] [CrossRef] [PubMed]
- Bernson, E.; Hallner, A.; Sander, F.E.; Wilsson, O.; Werlenius, O.; Rydström, A.; Kiffin, R.; Brune, M.; Foà, R.; Aurelius, J.; et al. Impact of killer-immunoglobulin-like receptor and human leukocyte antigen genotypes on the efficacy of immunotherapy in acute myeloid leukemia. Leukemia 2017, 31, 2552–2559. [Google Scholar] [CrossRef] [Green Version]
- Bryceson, Y.T.; March, M.E.; Barber, D.F.; Ljunggren, H.G.; Long, E.O. Cytolytic granule polarization and degranulation controlled by different receptors in resting NK cells. J. Exp. Med. 2005, 202, 1001–1012. [Google Scholar] [CrossRef]
- 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] [PubMed] [Green Version]
- 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]
- Rey, J.; Fauriat, C.; Kochbati, E.; Orlanducci, F.; Charbonnier, A.; D’Incan, E.; Andre, P.; Romagne, F.; Barbarat, B.; Vey, N.; et al. Kinetics of cytotoxic lymphocytes reconstitution after induction chemotherapy in elderly AML patients reveals progressive recovery of normal phenotypic and functional features in NK cells. Front. Immunol. 2017, 8, 64. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Mastaglio, S.; Wong, E.; Perera, T.; Ripley, J.; Blombery, P.; Smyth, M.J.; Koldej, R.; Ritchie, D. Natural killer receptor ligand expression on acute myeloid leukemia impacts survival and relapse after chemotherapy. Blood Adv. 2018, 2, 335–346. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Carlsten, M.; Järås, M. Natural Killer Cells in Myeloid Malignancies: Immune Surveillance, NK Cell Dysfunction, and Pharmacological Opportunities to Bolster the Endogenous NK Cells. Front. Immunol. 2019, 10, 2357. [Google Scholar] [CrossRef] [Green Version]
- Gill, S.; Vasey, A.E.; De Souza, A.; Baker, J.; Smith, A.T.; Kohrt, H.E.; Florek, M.; Gibbs, K.D.; Tate, K.; Ritchie, D.S.; et al. Rapid development of exhaustion and down-regulation of eomesodermin limit the antitumor activity of adoptively transferred murine natural killer cells. Blood 2012, 119, 5758–5768. [Google Scholar] [CrossRef]
- Wetzler, M.; Baer, M.R.; Stewart, S.J.; Donohue, K.; Ford, L.; Stewart, C.C.; Repasky, E.A.; Ferrone, S. HLA class I antigen cell surface expression is preserved on acute myeloid leukemia blasts at diagnosis and at relapse. Leukemia 2001, 15, 128–133. [Google Scholar] [CrossRef] [Green Version]
- Paczulla, A.M.; Rothfelder, K.; Raffel, S.; Konantz, M.; Steinbacher, J.; Wang, H.; Tandler, C.; Mbarga, M.; Schaefer, T.; Falcone, M.; et al. Absence of NKG2D ligands defines leukaemia stem cells and mediates their immune evasion. Nature 2019, 572, 254–259. [Google Scholar] [CrossRef]
- Baraganõ Raneros, A.; Martín-Palanco, V.; Fernandez, A.F.; Rodriguez, R.M.; Fraga, M.F.; Lopez-Larrea, C.; Suarez-Alvarez, B. Methylation of NKG2D ligands contributes to immune system evasion in acute myeloid leukemia. Genes Immun. 2015, 16, 71–82. [Google Scholar] [CrossRef] [PubMed]
- Coles, S.J.; Wang, E.C.Y.; Man, S.; Hills, R.K.; Burnett, A.K.; Tonks, A.; Darley, R.L. CD200 expression suppresses natural killer cell function and directly inhibits patient anti-tumor response in acute myeloid leukemia. Leukemia 2011, 25, 792–799. [Google Scholar] [CrossRef] [Green Version]
- Ho, J.M.; Dobson, S.M.; Voisin, V.; McLeod, J.; Kennedy, J.A.; Mitchell, A.; Jin, L.; Eppert, K.; Bader, G.; Minden, M.D.; et al. CD200 expression marks leukemia stem cells in human AML. Blood Adv. 2020, 4, 5402–5413. [Google Scholar] [CrossRef]
- Fauriat, C.; Just-Landi, S.; Mallet, F.; Arnoulet, C.; Sainty, D.; Olive, D.; Costello, R.T. Deficient expression of NCR in NK cells from acute myeloid leukemia: Evolution during leukemia treatment and impact of leukemia cells in NCR dull phenotype induction. Blood 2007, 109, 323–330. [Google Scholar] [CrossRef] [PubMed]
- Szczepanski, M.J.; Szajnik, M.; Welsh, A.; Whiteside, T.L.; Boyiadzis, M. Blast-derived microvesicles in sera from patients with acute myeloid leukemia suppress natural killer cell function via membrane-associated transforming growth factor-β1. Haematologica 2011, 96, 1302–1309. [Google Scholar] [CrossRef] [PubMed]
- Hilpert, J.; Grosse-Hovest, L.; Grünebach, F.; Buechele, C.; Nuebling, T.; Raum, T.; Steinle, A.; Salih, H.R. Comprehensive Analysis of NKG2D Ligand Expression and Release in Leukemia: Implications for NKG2D-Mediated NK Cell Responses. J. Immunol. 2012, 189, 1360–1371. [Google Scholar] [CrossRef] [Green Version]
- Teng, R.; Wang, Y.; Lv, N.; Zhang, D.; Williamson, R.A.; Lei, L.; Chen, P.; Lei, L.; Wang, B.; Fu, J.; et al. Hypoxia Impairs NK Cell Cytotoxicity through SHP-1-Mediated Attenuation of STAT3 and ERK Signaling Pathways. J. Immunol. Res. 2020, 2020, 4598476. [Google Scholar] [CrossRef] [PubMed]
- Stringaris, K.; Sekine, T.; Khoder, A.; Alsuliman, A.; Razzaghi, B.; Sargeant, R.; Pavlu, J.; Brisley, G.; de Lavallade, H.; Sarvaria, A.; et al. Leukemia-induced phenotypic and functional defects in natural killer cells predict failure to achieve remission in acute myeloid leukemia. Haematologica 2014, 99, 836–847. [Google Scholar] [CrossRef] [Green Version]
- Nguyen, S.; Beziat, V.; Dhedin, N.; Kuentz, M.; Vernant, J.P.; Debre, P.; Vieillard, V. HLA-E upregulation on IFN-γ-activated AML blasts impairs CD94/ NKG2A-dependent NK cytolysis after haplo-mismatched hematopoietic SCT. Bone Marrow Transplant. 2009, 43, 693–699. [Google Scholar] [CrossRef] [Green Version]
- Kearney, C.J.; Ramsbottom, K.M.; Voskoboinik, I.; Darcy, P.K.; Oliaro, J. Loss of DNAM-1 ligand expression by acute myeloid leukemia cells renders them resistant to NK cell killing. Oncoimmunology 2016, 5. [Google Scholar] [CrossRef] [Green Version]
- Valhondo, I.; Hassouneh, F.; Lopez-Sejas, N.; Pera, A.; Sanchez-Correa, B.; Guerrero, B.; Bergua, J.M.; Arcos, M.J.; Bañas, H.; Casas-Avilés, I.; et al. Characterization of the DNAM-1, TIGIT and TACTILE axis on circulating NK, NKT-like and T cell subsets in patients with acute myeloid leukemia. Cancers 2020, 12, 2171. [Google Scholar] [CrossRef] [PubMed]
- Wisnovsky, S.; Möckl, L.; Malaker, S.A.; Pedram, K.; Hess, G.T.; Riley, N.M.; Gray, M.A.; Smith, B.A.H.; Bassik, M.C.; Moerner, W.E.; et al. Genome-wide CRISPR screens reveal a specific ligand for the glycan-binding immune checkpoint receptor Siglec-7. Proc. Natl. Acad. Sci. USA 2021, 118. [Google Scholar] [CrossRef]
- Gillissen, M.A.; De Jong, G.; Kedde, M.; Yasuda, E.; Levie, S.E.; Moiset, G.; Hensbergen, P.J.; Bakker, A.Q.; Wagner, K.; Villaudy, J.; et al. Patient-derived antibody recognizes a unique CD43 epitope expressed on all AML and has antileukemia activity in mice. Blood Adv. 2017, 1, 1551–1564. [Google Scholar] [CrossRef] [Green Version]
- Parameswaran, R.; Ramakrishnan, P.; Moreton, S.A.; Xia, Z.; Hou, Y.; Lee, D.A.; Gupta, K.; Delima, M.; Beck, R.C.; Wald, D.N. Repression of GSK3 restores NK cell cytotoxicity in AML patients. Nat. Commun. 2016, 7, 1–11. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Schmiedel, B.J.; Nuebling, T.; Steinbacher, J.; Malinovska, A.; Wende, C.M.; Azuma, M.; Schneider, P.; Grosse-Hovest, L.; Salih, H.R. Receptor Activator for NF-κB Ligand in Acute Myeloid Leukemia: Expression, Function, and Modulation of NK Cell Immunosurveillance. J. Immunol. 2013, 190, 821–831. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Peterlin, P.; Gaschet, J.; Guillaume, T.; Garnier, A.; Eveillard, M.; Le Bourgeois, A.; Cherel, M.; Debord, C.; Le Bris, Y.; Theisen, O.; et al. A new cytokine-based dynamic stratification during induction is highly predictive of survivals in acute myeloid leukemia. Cancer Med. 2021, 10, 642–648. [Google Scholar] [CrossRef]
- Wu, J.; Gao, F.X.; Wang, C.; Qin, M.; Han, F.; Xu, T.; Hu, Z.; Long, Y.; He, X.M.; Deng, X.; et al. IL-6 and IL-8 secreted by tumour cells impair the function of NK cells via the STAT3 pathway in oesophageal squamous cell carcinoma. J. Exp. Clin. Cancer Res. 2019, 38, 321. [Google Scholar] [CrossRef]
- Chan, L.C.; Li, C.W.; Xia, W.; Hsu, J.M.; Lee, H.H.; Cha, J.H.; Wang, H.L.; Yang, W.H.; Yen, E.Y.; Chang, W.C.; et al. IL-6/JAK1 pathway drives PD-L1 Y112 phosphorylation to promote cancer immune evasion. J. Clin. Invest. 2019, 129, 3324–3338. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Mundy-Bosse, B.L.; Scoville, S.D.; Chen, L.; McConnell, K.; Mao, H.C.; Ahmed, E.H.; Zorko, N.; Harvey, S.; Cole, J.; Zhang, X.; et al. MicroRNA-29b mediates altered innate immune development in acute leukemia. J. Clin. Invest. 2016, 126, 4404–4416. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Scoville, S.D.; Nalin, A.P.; Chen, L.; Chen, L.; Zhang, M.H.; McConnell, K.; Beceiro Casas, S.; Ernst, G.; Al-Rahman Traboulsi, A.; Hashi, N.; et al. Human AML activates the aryl hydrocarbon receptor pathway to impair NK cell development and function. Blood 2018, 132, 1792–1804. [Google Scholar] [CrossRef] [PubMed]
- Aurelius, J.; Thorén, F.B.; Akhiani, A.A.; Brune, M.; Palmqvist, L.; Hansson, M.; Hellstrand, K.; Martner, A. Monocytic AML cells inactivate antileukemic lymphocytes: Role of NADPH oxidase/gp91 phox expression and the PARP-1/PAR pathway of apoptosis. Blood 2012, 119, 5832–5837. [Google Scholar] [CrossRef] [Green Version]
- Xiao, G.; Wang, X.; Sheng, J.; Lu, S.; Yu, X.; Wu, J.D. Soluble NKG2D ligand promotes MDSC expansion and skews macrophage to the alternatively activated phenotype. J. Hematol. Oncol. 2015, 8, 13. [Google Scholar] [CrossRef] [Green Version]
- Curti, A.; Trabanelli, S.; Salvestrini, V.; Baccarani, M.; Lemoli, R.M. The role of indoleamine 2,3-dioxygenase in the induction of immune tolerance: Focus on hematology. Blood 2009, 113, 2394–2401. [Google Scholar] [CrossRef] [PubMed]
- Ghiringhelli, F.; Ménard, C.; Terme, M.; Flament, C.; Taieb, J.; Chaput, N.; Puig, P.E.; Novault, S.; Escudier, B.; Vivier, E.; et al. CD4+CD25+ regulatory T cells inhibit natural killer cell functions in a transforming growth factor-β-dependent manner. J. Exp. Med. 2005, 202, 1075–1085. [Google Scholar] [CrossRef] [PubMed]
- Sarhan, D.; Cichocki, F.; Zhang, B.; Yingst, A.; Spellman, S.R.; Cooley, S.; Verneris, M.R.; Blazar, B.R.; Miller, J.S. Adaptive NK cells with low TIGIT expression are inherently resistant to myeloid-derived suppressor cells. Cancer Res. 2016, 76, 5696–5706. [Google Scholar] [CrossRef] [Green Version]
- 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]
- Ni, J.; Wang, X.; Stojanovic, A.; Zhang, Q.; Wincher, M.; Bühler, L.; Arnold, A.; Correia, M.P.; Winkler, M.; Koch, P.S.; et al. Single-Cell RNA Sequencing of Tumor-Infiltrating NK Cells Reveals that Inhibition of Transcription Factor HIF-1α Unleashes NK Cell Activity. Immunity 2020, 52, 1075–1087.e8. [Google Scholar] [CrossRef] [PubMed]
- Sweeney, C.; Vyas, P. The Graft-Versus-Leukemia Effect in AML. Front. Oncol. 2019, 9, 1217. [Google Scholar] [CrossRef]
- Loke, J.; Malladi, R.; Moss, P.; Craddock, C. The role of allogeneic stem cell transplantation in the management of acute myeloid leukaemia: A triumph of hope and experience. Br. J. Haematol. 2020, 188, 129–146. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Maggs, L.; Kinsella, F.; Tracey Chan, Y.L.; Eldershaw, S.; Murray, D.; Nunnick, J.; Bird, J.; Craddock, C.; Zuo, J.; Malladi, R.; et al. The number of CD56dim NK cells in the graft has a major impact on risk of disease relapse following allo-HSCT. Blood Adv. 2017, 1, 1589–1597. [Google Scholar] [CrossRef] [Green Version]
- Ruggeri, L.; Mancusi, A.; Capanni, M.; Urbani, E.; Carotti, A.; Aloisi, T.; Stern, M.; Pende, D.; Perruccio, K.; Burchielli, E.; et al. Donor natural killer cell allorecognition of missing self in haploidentical hematopoietic transplantation for acute myeloid leukemia: Challenging its predictive value. Blood 2007, 110, 433–440. [Google Scholar] [CrossRef] [Green Version]
- Venstrom, J.M.; Pittari, G.; Gooley, T.A.; Chewning, J.H.; Spellman, S.; Haagenson, M.; Gallagher, M.M.; Malkki, M.; Petersdorf, E.; Dupont, B.; et al. HLA-C –Dependent Prevention of Leukemia Relapse by Donor Activating KIR2DS1. N. Engl. J. Med. 2012, 367, 805–816. [Google Scholar] [CrossRef] [Green Version]
- Cooley, S.; Weisdorf, D.J.; Guethlein, L.A.; Klein, J.P.; Wang, T.; Marsh, S.G.E.; Spellman, S.; Haagenson, M.D.; Saeturn, K.; Ladner, M.; et al. Donor Killer Cell Ig-like Receptor B Haplotypes, Recipient HLA-C1, and HLA-C Mismatch Enhance the Clinical Benefit of Unrelated Transplantation for Acute Myelogenous Leukemia. J. Immunol. 2014, 192, 4592–4600. [Google Scholar] [CrossRef]
- Shaffer, B.C.; Hsu, K.C. How important is NK alloreactivity and KIR in allogeneic transplantation? Best Pract. Res. Clin. Haematol. 2016, 29, 351–358. [Google Scholar] [CrossRef]
- Rosenberg, S.A.; Lotze, M.T.; Muul, L.M.; Chang, A.E.; Avis, F.P.; Leitman, S.; Linehan, W.M.; Robertson, C.N.; Lee, R.E.; Rubin, J.T.; et al. A Progress Report on the Treatment of 157 Patients with Advanced Cancer Using Lymphokine-Activated Killer Cells and Interleukin-2 or High-Dose Interleukin-2 Alone. N. Engl. J. Med. 1987, 316, 889–897. [Google Scholar] [CrossRef]
- Dudley, M.E.; Wunderlich, J.R.; Robbins, P.F.; Yang, J.C.; Hwu, P.; Schwartzentruber, D.J.; Topalian, S.L.; Sherry, R.; Restifo, N.P.; Hubicki, A.M.; et al. Cancer regression and autoimmunity in patients after clonal repopulation with antitumor lymphocytes. Science 2002, 298, 850–854. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Miller, J.S.; Lanier, L.L. Natural killer cells in cancer immunotherapy. Annu. Rev. Cancer Biol. 2019, 3, 77–103. [Google Scholar] [CrossRef] [Green Version]
- Bachanova, V.; Cooley, S.; Defor, T.E.; Verneris, M.R.; Zhang, B.; McKenna, D.H.; Curtsinger, J.; Panoskaltsis-Mortari, A.; Lewis, D.; Hippen, K.; et al. Clearance of acute myeloid leukemia by haploidentical natural killer cells is improved using IL-2 diphtheria toxin fusion protein. Blood 2014, 123, 3855–3863. [Google Scholar] [CrossRef] [PubMed]
- Cooley, S.; He, F.; Bachanova, V.; Vercellotti, G.M.; DeFor, T.E.; Curtsinger, J.M.; Robertson, P.; Grzywacz, B.; Conlon, K.C.; Waldmann, T.A.; et al. First-in-human trial of rhIL-15 and haploidentical natural killer cell therapy for advanced acute myeloid leukemia. Blood Adv. 2019, 3, 1970–1980. [Google Scholar] [CrossRef] [PubMed]
- Curti, A.; Ruggeri, L.; Parisi, S.; Bontadini, A.; Dan, E.; Motta, M.R.; Rizzi, S.; Trabanelli, S.; Ocadlikova, D.; Lecciso, M.; et al. Cancer Therapy: Clinical Larger Size of Donor Alloreactive NK Cell Repertoire Correlates with Better Response to NK Cell Immunotherapy in Elderly Acute Myeloid Leukemia Patients. Clin Cancer Res 2016, 22. [Google Scholar] [CrossRef] [Green Version]
- Björklund, A.T.; Carlsten, M.; Sohlberg, E.; Liu, L.L.; Clancy, T.; Karimi, M.; Cooley, S.; Miller, J.S.; Klimkowska, M.; Schaffer, M.; et al. Complete Remission with Reduction of High-Risk Clones following Haploidentical NK-Cell Therapy against MDS and AML. Clin. Cancer Res. 2018, 24, 1834–1844. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Rubnitz, J.E.; Inaba, H.; Ribeiro, R.C.; Pounds, S.; Rooney, B.; Bell, T.; Pui, C.H.; Leung, W. NKAML: A pilot study to determine the safety and feasibility of haploidentical natural killer cell transplantation in childhood acute myeloid leukemia. J. Clin. Oncol. 2010, 28, 955–959. [Google Scholar] [CrossRef] [Green Version]
- Nguyen, R.; Wu, H.; Pounds, S.; Inaba, H.; Ribeiro, R.C.; Cullins, D.; Rooney, B.; Bell, T.; Lacayo, N.J.; Heym, K.; et al. A phase II clinical trial of adoptive transfer of haploidentical natural killer cells for consolidation therapy of pediatric acute myeloid leukemia. J. Immunother. Cancer 2019, 7, 81. [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]
- Dolstra, H.; Roeven, M.W.H.; Spanholtz, J.; Hangalapura, B.N.; Tordoir, M.; Maas, F.; Leenders, M.; Bohme, F.; Kok, N.; Trilsbeek, C.; et al. Successful transfer of umbilical cord blood CD34+ hematopoietic stem and progenitor-derived NK cells in older acute myeloid leukemia patients. Clin. Cancer Res. 2017, 23, 4107–4118. [Google Scholar] [CrossRef] [Green Version]
- Basar, R.; Daher, M.; Rezvani, K. Next-generation cell therapies: The emerging role of CAR-NK cells. Blood Adv. 2020, 4, 5868–5876. [Google Scholar] [CrossRef] [PubMed]
- Zhang, C.; Hu, Y.; Shi, C. Targeting Natural Killer Cells for Tumor Immunotherapy. Front. Immunol. 2020, 11, 60. [Google Scholar] [CrossRef] [Green Version]
- Kerbauy, L.N.; Ang, S.; Liu, E.; Banerjee, P.P.; Wu, Y.; Shaim, H.; Lim, F.L.W.I.; Basar, R.; Li, L.; Muftuoglu, M.; et al. Cord Blood NK Cells Engineered to Express a Humanized CD123-Targeted Chimeric Antigen Receptor (CAR) and IL-15 As Off-the-Shelf Therapy for Acute Myeloid Leukemia. Blood 2017, 130, 4453. [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] [Green Version]
- Shankar, K.; Capitini, C.M.; Capitini, C.M.; Saha, K.; Saha, K.; Saha, K. Genome engineering of induced pluripotent stem cells to manufacture natural killer cell therapies. Stem Cell Res. Ther. 2020, 11, 1–14. [Google Scholar] [CrossRef] [PubMed]
- Hermanson, D.L.; Bendzick, L.; Pribyl, L.; McCullar, V.; Vogel, R.I.; Miller, J.S.; Geller, M.A.; Kaufman, D.S. Induced Pluripotent Stem Cell-Derived Natural Killer Cells for Treatment of Ovarian Cancer. Stem Cells 2016, 34, 93–101. [Google Scholar] [CrossRef] [Green Version]
- Bachanova, V.; Cayci, Z.; Lewis, D.; Maakaron, J.E.; Janakiram, M.; Bartz, A.; Payne, S.; Wong, C.; Cooley, S.; Valamehr, B.; et al. Initial Clinical Activity of FT596, a First-in-Class, Multi-Antigen Targeted, Off-the-Shelf, iPSC-Derived CD19 CAR NK Cell Therapy in Relapsed/Refractory B-Cell Lymphoma. Blood 2020, 136, 8. [Google Scholar] [CrossRef]
- McLellan, A.D.; Ali Hosseini Rad, S.M. Chimeric antigen receptor T cell persistence and memory cell formation. Immunol. Cell Biol. 2019, 97, 664–674. [Google Scholar] [CrossRef]
- Turtle, C.J.; Hanafi, L.A.; Berger, C.; Gooley, T.A.; Cherian, S.; Hudecek, M.; Sommermeyer, D.; Melville, K.; Pender, B.; Budiarto, T.M.; et al. CD19 CAR-T cells of defined CD4+:CD8+ composition in adult B cell ALL patients. J. Clin. Invest. 2016, 126, 2123–2138. [Google Scholar] [CrossRef] [Green Version]
- Abel, A.M.; Yang, C.; Thakar, M.S.; Malarkannan, S. Natural killer cells: Development, maturation, and clinical utilization. Front. Immunol. 2018, 9, 1. [Google Scholar] [CrossRef] [Green Version]
- Oei, V.Y.S.; Siernicka, M.; Graczyk-Jarzynka, A.; Hoel, H.J.; Yang, W.; Palacios, D.; Sbak, H.A.; 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]
- Gang, M.; Marin, N.D.; Wong, P.; Neal, C.C.; Marsala, L.; Foster, M.; Schappe, T.; Meng, W.; Tran, J.; Schaettler, M.; et al. CAR-modified memory-like NK cells exhibit potent responses to NK-resistant lymphomas. Blood 2020, 136, 2308–2318. [Google Scholar] [CrossRef]
- Li, C.; Yang, N.; Li, H.; Wang, Z. Robo1-specific chimeric antigen receptor natural killer cell therapy for pancreatic ductal adenocarcinoma with liver metastasis. J. Cancer Res. Ther. 2020, 16, 393. [Google Scholar] [CrossRef]
- 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]
- Ehninger, A.; Kramer, M.; Röllig, C.; Thiede, C.; Bornhäuser, M.; Von Bonin, M.; Wermke, M.; Feldmann, A.; Bachmann, M.; Ehninger, G.; et al. Distribution and levels of cell surface expression of CD33 and CD123 in acute myeloid leukemia. Blood Cancer J. 2014, 4, e218. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Kararoudi, M.N.; Likhite, S.; Elmas, E.; Schwartz, M.; Sorathia, K.; Yamamoto, K.; Chakravarti, N.; Moriarity, B.S.; Meyer, K.; Lee, D.A. CD33 Targeting Primary CAR-NK Cells Generated By CRISPR Mediated Gene Insertion Show Enhanced Anti-AML Activity. Blood 2020, 136, 3. [Google Scholar] [CrossRef]
- Sinha, C.; Seth, A.; Kahali, B.; Cunningham, L. Development and Evaluation of NK-CD123 CAR Against High Risk Acute Myeloid Leukemia. Biol. Blood Marrow Transplant. 2017, 23, S253. [Google Scholar] [CrossRef] [Green Version]
- Kloess, S.; Oberschmidt, O.; Dahlke, J.; Vu, X.-K.; Neudoerfl, C.; Kloos, A.; Gardlowski, T.; Matthies, N.; Heuser, M.; Meyer, J.; et al. Preclinical Assessment of Suitable Natural Killer Cell Sources for Chimeric Antigen Receptor Natural Killer–Based “Off-the-Shelf” Acute Myeloid Leukemia Immunotherapies. Hum. Gene Ther. 2019, 30, 381–401. [Google Scholar] [CrossRef]
- Christodoulou, I.; Marple, A.; Ravich, W.; Bonifant, C. CD123-CAR NK Cells are Activated by and Kill Acute Myeloid Leukemia. In Proceedings of the American Society of Gene and Cell Therapy 23rd Annual Meeting, Boston, MA, USA, 12–15 May 2020; pp. 150–151. [Google Scholar]
- Wang, J.; Chen, S.; Xiao, W.; Li, W.; Wang, L.; Yang, S.; Wang, W.; Xu, L.; Liao, S.; Liu, W.; et al. CAR-T cells targeting CLL-1 as an approach to treat acute myeloid leukemia. J. Hematol. Oncol. 2018, 11, 7. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Leivas, A.; Rio, P.; Mateos, R.; Paciello, M.L.; Garcia-Ortiz, A.; Fernandez, L.; Perez-Martinez, A.; Lee, D.A.; Powell, D.J.; Valeri, A.; et al. NKG2D-CAR Transduced Primary Natural Killer Cells Efficiently Target Multiple Myeloma Cells. Blood 2018, 132, 590. [Google Scholar] [CrossRef]
- Gurney, M.; Stikvoort, A.; Nolan, E.; Kirkham-Mccarthy, L.; Khoruzhenko, S.; Shivakumar, R.; Zweegman, S.; Van De Donk, N.W.C.J.; Mutis, T.; Szegezdi, E.; et al. CD38 knockout natural killer cells expressing an affinity optimized CD38 chimeric antigen receptor successfully target acute myeloid leukemia with reduced effector cell fratricide. Haematologica 2020, 105. [Google Scholar] [CrossRef]
- Dong, H.; Xie, G.; Liang, Y.; Dongjoo Ham, J.; Vergara, J.; Chen, J.; Ritz, J.; Romee, R. Engineered Memory-like NK Cars Targeting a Neoepitope Derived from Intracellular NPM1c Exhibit Potent Activity and Specificity Against Acute Myeloid Leukemia. Blood 2020, 136, 3–4. [Google Scholar] [CrossRef]
- Del Poeta, G.; Stasi, R.; Venditti, A.; Cox, C.; Aronica, G.; Masi, M.; Bruno, A.; Simone, M.D.; Buccisano, F.; Papa, G. CD7 expression in acute myeloid leukemia. Leuk. Lymphoma 1995, 17, 111–119. [Google Scholar] [CrossRef]
- 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]
- Eckel, A.M.; Cherian, S.; Miller, V.; Soma, L. CD33 expression on natural killer cells is a potential confounder for residual disease detection in acute myeloid leukemia by flow cytometry. Cytom. Part B - Clin. Cytom. 2020, 98, 174–178. [Google Scholar] [CrossRef]
- Kloess, S.; da Silva, A.E.V.; Oberschmidt, O.; Gardlowski, T.; Matthies, N.; Vyas, M.; Arseniev, L.; Heuser, M.; von Strandmann, E.P.; Köhl, U. Triplebody mediates increased anti-leukemic reactivity of IL-2 activated donor natural killer (NK) cells and impairs viability of their CD33-expressing NK subset. Front. Immunol. 2017, 8, 1100. [Google Scholar] [CrossRef] [Green Version]
- Klöß, S.; Oberschmidt, O.; Morgan, M.; Dahlke, J.; Arseniev, L.; Huppert, V.; Granzin, M.; Gardlowski, T.; Matthies, N.; Soltenborn, S.; et al. Optimization of Human NK Cell Manufacturing: Fully Automated Separation, Improved Ex Vivo Expansion Using IL-21 with Autologous Feeder Cells, and Generation of Anti-CD123-CAR-Expressing Effector Cells. Hum. Gene Ther. 2017, 28, 897–913. [Google Scholar] [CrossRef] [PubMed]
- Kararoudi, M.N.; Nagai, Y.; Elmas, E.; de Souza Fernandes Pereira, M.; Ali, S.A.; Imus, P.H.; Wethington, D.; Borrello, I.M.; Lee, D.A.; Ghiaur, G. CD38 deletion of human primary NK cells eliminates daratumumab-induced fratricide and boosts their effector activity. Blood 2020, 136, 2416–2427. [Google Scholar] [CrossRef] [PubMed]
- Cichocki, F.; Woan, K.; Wu, C.-Y.; Blazar, B.R.; Bjordahl, R.; Valamehr, B.; Miller, J.S. NK Cells Lacking CD38 Are Resistant to Oxidative Stress-Induced Death. Blood 2019, 134, 3215. [Google Scholar] [CrossRef]
- Chang, Y.H.; Connolly, J.; Shimasaki, N.; Mimura, K.; Kono, K.; Campana, D. A chimeric receptor with NKG2D specificity enhances natural killer cell activation and killing of tumor cells. Cancer Res. 2013, 73, 1777–1786. [Google Scholar] [CrossRef] [Green Version]
- Maroto-Martin, E. NKG2D and BCMA-CAR NK Cells Efficiently Eliminate Multiple Myeloma Cells. A Comprehensive Comparison between Two Clinically Relevant CARs. In Proceedings of the EHA 24th Congress, Amsterdam, The Netherlands, 13–16 June 2019. [Google Scholar]
- Fultang, L.; Panetti, S.; Ng, M.; Collins, P.; Graef, S.; Rizkalla, N.; Booth, S.; Lenton, R.; Noyvert, B.; Shannon-Lowe, C.; et al. MDSC targeting with Gemtuzumab ozogamicin restores T cell immunity and immunotherapy against cancers. EBioMedicine 2019. [Google Scholar] [CrossRef] [Green Version]
- Krejcik, J.; Casneuf, T.; Nijhof, I.S.; Verbist, B.; Bald, J.; Plesner, T.; Syed, K.; Liu, K.; Van De Donk, N.W.C.J.; Weiss, B.M.; et al. Daratumumab depletes CD38+ immune regulatory cells, promotes T-cell expansion, and skews T-cell repertoire in multiple myeloma. Blood 2016, 128, 384–394. [Google Scholar] [CrossRef] [Green Version]
- Barber, A.; Rynda, A.; Sentman, C.L. Chimeric NKG2D Expressing T Cells Eliminate Immunosuppression and Activate Immunity within the Ovarian Tumor Microenvironment. J. Immunol. 2009, 183, 6939–6947. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Daher, M.; Basar, R.; Gokdemir, E.; Baran, N.; Uprety, N.; Nunez Cortes, A.K.; Mendt, M.; Kerbauy, L.N.; Banerjee, P.P.; Hernandez Sanabria, M.; et al. Targeting a cytokine checkpoint enhances the fitness of armored cord blood CAR-NK cells. Blood 2020, 137, 624–636. [Google Scholar] [CrossRef]
- Zhu, H.; Blum, R.; Wu, Z.; Meng, Z.; Guan, K.-L.; Kaufman, D.S. Deletion of CISH in Human Pluripotent Stem Cell-Derived Natural Killer Cells Enhances Anti-Tumor Activity Via Metabolic Reprogramming. Blood 2019, 134, 619. [Google Scholar] [CrossRef]
- Wang, X.; Jasinski, D.L.; Medina, J.L.; Spencer, D.M.; Foster, A.E.; Bayle, J.H. Inducible MyD88/CD40 synergizes with IL-15 to enhance antitumor efficacy of CAR-NK cells Key Points. Blood Adv. 2020, 12, 1950–1964. [Google Scholar] [CrossRef] [PubMed]
- Collinson-Pautz, M.R.; Chang, W.C.; Lu, A.; Khalil, M.; Crisostomo, J.W.; Lin, P.Y.; Mahendravada, A.; Shinners, N.P.; Brandt, M.E.; Zhang, M.; et al. Constitutively active MyD88/CD40 costimulation enhances expansion and efficacy of chimeric antigen receptor T cells targeting hematological malignancies. Leukemia 2019, 33, 2195–2207. [Google Scholar] [CrossRef] [Green Version]
- Uppendahl, L.D.; Felices, M.; Bendzick, L.; Ryan, C.; Kodal, B.; Hinderlie, P.; Boylan, K.L.M.; Skubitz, A.P.N.; Miller, J.S.; Geller, M.A. Cytokine-induced memory-like natural killer cells have enhanced function, proliferation, and in vivo expansion against ovarian cancer cells. Gynecol. Oncol. 2019, 153, 149–157. [Google Scholar] [CrossRef]
- Grzywacz, B.; Moench, L.; McKenna, D.; Tessier, K.M.; Bachanova, V.; Cooley, S.; Miller, J.S.; Courville, E.L. Natural Killer Cell Homing and Persistence in the Bone Marrow after Adoptive Immunotherapy Correlates with Better Leukemia Control. J. Immunother. 2019, 42, 65–72. [Google Scholar] [CrossRef]
- Levy, E.; Reger, R.; Segerberg, F.; Lambert, M.; Leijonhufvud, C.; Baumer, Y.; Carlsten, M.; Childs, R. Enhanced Bone Marrow Homing of Natural Killer Cells Following mRNA Transfection With Gain-of-Function Variant CXCR4R334X. Front. Immunol. 2019, 10, 1262. [Google Scholar] [CrossRef] [Green Version]
- Barbier, V.; Erbani, J.; Fiveash, C.; Davies, J.M.; Tay, J.; Tallack, M.R.; Lowe, J.; Magnani, J.L.; Pattabiraman, D.R.; Perkins, A.C.; et al. Endothelial E-selectin inhibition improves acute myeloid leukaemia therapy by disrupting vascular niche-mediated chemoresistance. Nat. Commun. 2020, 11. [Google Scholar] [CrossRef]
- Sackstein, R. The First Step in Adoptive Cell Immunotherapeutics: Assuring Cell Delivery via Glycoengineering. Front. Immunol. 2019, 9, 3084. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Hong, S.; Yu, C.; Wang, P.; Shi, Y.; Cheng, B.; Chen, M.; Chapla, D.; Reigh, N.; Narimatsu, Y.; Chen, X.; et al. Glycoengineering of NK cells with glycan ligands of CD22 and selectins for B-cell lymphoma therapy. bioRxiv 2020. [Google Scholar] [CrossRef]
- Khan, M.; Arooj, S.; Wang, H. NK Cell-Based Immune Checkpoint Inhibition. Front. Immunol. 2020, 11, 167. [Google Scholar] [CrossRef] [PubMed]
- Pesce, S.; Trabanelli, S.; Di Vito, C.; Greppi, M.; Obino, V.; Guolo, F.; Minetto, P.; Bozzo, M.; Calvi, M.; Zaghi, E.; et al. Cancer immunotherapy by blocking immune checkpoints on innate lymphocytes. Cancers 2020, 12, 3504. [Google Scholar] [CrossRef]
- Vey, N.; Dumas, P.-Y.; Recher, C.; Gastaud, L.; Lioure, B.; Bulabois, C.-E.; Pautas, C.; Marolleau, J.-P.; Leprêtre, S.; Raffoux, E.; et al. Randomized Phase 2 Trial of Lirilumab (anti-KIR monoclonal antibody, mAb) As Maintenance Treatment in Elderly Patients (pts) with Acute Myeloid Leukemia (AML): Results of the Effikir Trial. Blood 2017, 130, 889. [Google Scholar] [CrossRef]
- Roberto, A.; Di Vito, C.; Zaghi, E.; Mazza, E.M.C.; Capucetti, A.; Calvi, M.; Tentorio, P.; Zanon, V.; Sarina, B.; Mariotti, J.; et al. The early expansion of anergic NKG2A pos /CD56 dim /CD16 neg natural killer represents a therapeutic target in haploidentical hematopoietic stem cell transplantation. Haematologica 2018, 103, 1390–1402. [Google Scholar] [CrossRef] [Green Version]
- Ruggeri, L.; Urbani, E.; André, P.; Mancusi, A.; Tosti, A.; Topini, F.; Bléry, M.; Animobono, L.; Romagné, F.; Wagtmann, N.; et al. Effects of anti-NKG2A antibody administration on leukemia and normal hematopoietic cells. Haematologica 2016, 101, 626–633. [Google Scholar] [CrossRef] [Green Version]
- Mahaweni, N.M.; Ehlers, F.A.I.; Sarkar, S.; Janssen, J.W.H.; Tilanus, M.G.J.; Bos, G.M.J.; Wieten, L. NKG2A Expression Is Not per se Detrimental for the Anti-Multiple Myeloma Activity of Activated Natural Killer Cells in an In Vitro System Mimicking the Tumor Microenvironment. Front. Immunol. 2018, 9, 22. [Google Scholar] [CrossRef]
- Kamiya, T.; Seow, S.V.; Wong, D.; Robinson, M.; Campana, D. Blocking expression of inhibitory receptor NKG2A overcomes tumor resistance to NK cells. J. Clin. Invest. 2019, 129, 2094–2106. [Google Scholar] [CrossRef] [Green Version]
- Berger, R.; Rotem-Yehudar, R.; Slama, G.; Landes, S.; Kneller, A.; Leiba, M.; Koren-Michowitz, M.; Shimoni, A.; Nagler, A. Phase I Safety and Pharmacokinetic Study of CT-011, a Humanized Antibody Interacting with PD-1, in Patients with Advanced Hematologic Malignancies. Clin. Cancer Res. 2008, 14, 3044–3051. [Google Scholar] [CrossRef] [Green Version]
- Daver, N.G.; Garcia-Manero, G.; Konopleva, M.Y.; Alfayez, M.; Pemmaraju, N.; Kadia, T.M.; DiNardo, C.D.; Cortes, J.E.; Ravandi, F.; Abbas, H.; et al. Azacitidine (AZA) with Nivolumab (Nivo), and AZA with Nivo + Ipilimumab (Ipi) in Relapsed/Refractory Acute Myeloid Leukemia: A Non-Randomized, Prospective, Phase 2 Study. Blood 2019, 134, 830. [Google Scholar] [CrossRef]
- Davis, Z.; Felices, M.; Lenvik, T.R.; Badal, S.; Hinderlie, P.; Blazar, B.R.; Vallera, D.A.; Miller, J.S. PD-1 Is Expressed at Low Levels on All Peripheral Blood Natural Killer Cells but Is a Significant Suppressor of NK Function Against PD-1 Ligand Expressing Tumor Targets. Blood 2019, 134, 621. [Google Scholar] [CrossRef]
- Jia, B.; Zhao, C.; Claxton, D.F.; Ehmann, W.C.; Rybka, W.B.; Mineishi, S.; Naik, S.; Songdej, N.; Khawaja, M.; Hohl, R.J.; et al. TIGIT Expression Positively Associates with NK Cell Function in AML Patients. Blood 2018, 132, 5250. [Google Scholar] [CrossRef]
- Rastogi, N.; Baker, S.; Man, S.; Uger, R.A.; Wong, M.; Coles, S.J.; Hodges, M.; Gilkes, A.F.; Knapper, S.; Darley, R.L.; et al. Use of an anti-CD200-blocking antibody improves immune responses to AML in vitro and in vivo. Br. J. Haematol. 2020, bjh.17125. [Google Scholar] [CrossRef] [PubMed]
- Herbrich, S.; Baggerly, K.; Alatrash, G.; Davis, R.E.; Andreeff, M.; Konopleva, M.Y. CD200 Is a Stem Cell-Specific Immunosuppressive Target in AML. Blood 2018, 132, 2768. [Google Scholar] [CrossRef]
- Huang, C.H.; Liao, Y.J.; Fan, T.H.; Chiou, T.J.; Lin, Y.H.; Twu, Y.C. A developed NK-92MI cell line with siglec-7neg phenotype exhibits high and sustainable cytotoxicity against leukemia cells. Int. J. Mol. Sci. 2018, 19, 1073. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Rohner, A.; Langenkamp, U.; Siegler, U.; Kalberer, C.P.; Wodnar-Filipowicz, A. Differentiation-promoting drugs up-regulate NKG2D ligand expression and enhance the susceptibility of acute myeloid leukemia cells to natural killer cell-mediated lysis. Leuk. Res. 2007, 31, 1393–1402. [Google Scholar] [CrossRef] [PubMed]
- Yun, H.D.; Schirm, D.K.; Felices, M.; Miller, J.S.; Eckfeldt, C.E. Dinaciclib enhances natural killer cell cytotoxicity against acute myelogenous leukemia. Blood Adv. 2019, 3, 2448–2452. [Google Scholar] [CrossRef] [PubMed]
- Zhu, S.; Denman, C.J.; Cobanoglu, Z.S.; Kiany, S.; Lau, C.C.; Gottschalk, S.M.; Hughes, D.P.M.; Kleinerman, E.S.; Lee, D.A. The narrow-spectrum HDAC inhibitor entinostat enhances NKG2D expression without NK cell toxicity, leading to enhanced recognition of cancer cells. Pharm. Res. 2015, 32, 779–792. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Poggi, A.; Catellani, S.; Garuti, A.; Pierri, I.; Gobbi, M.; Zocchi, M.R. Effective in vivo induction of NKG2D ligands in acute myeloid leukaemias by all-trans-retinoic acid or sodium valproate. Leukemia 2009, 23, 641–648. [Google Scholar] [CrossRef]
- Meng, X.W.; Koh, B.D.; Zhang, J.S.; Flatten, K.S.; Schneider, P.A.; Billadeau, D.D.; Hess, A.D.; Smith, B.D.; Karp, J.E.; Kaufmann, S.H. Poly(ADP-ribose) polymerase inhibitors sensitize cancer cells to death receptor-mediated apoptosis by enhancing death receptor expression. J. Biol. Chem. 2014, 289, 20543–20558. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Le Roy, A.; Prébet, T.; Castellano, R.; Goubard, A.; Riccardi, F.; Fauriat, C.; Granjeaud, S.; Benyamine, A.; Castanier, C.; Orlanducci, F.; et al. Immunomodulatory drugs exert anti-leukemia effects in acute myeloid leukemia by direct and immunostimulatory activities. Front. Immunol. 2018, 9. [Google Scholar] [CrossRef]
- Lehmann, C.; Zeis, M.; Schmitz, N.; Uharek, L. Impaired binding of perforin on the surface of tumor cells is a cause of target cell resistance against cytotoxic effector cells. Blood 2000, 96, 594–600. [Google Scholar] [CrossRef]
- Fritsch, K.; Finke, J.; Grüllich, C. Suppression of granzyme B activity and caspase-3 activation in leukaemia cells constitutively expressing the protease inhibitor 9. Ann. Hematol. 2013, 92, 1603–1609. [Google Scholar] [CrossRef]
- 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]
- Szegezdi, E.; Reis, C.R.; Der Sloot, A.M.V.; Natoni, A.; O’Reilly, A.; Reeve, J.; Cool, R.H.; O’Dwyer, M.; Knapper, S.; Serrano, L.; et al. Targeting AML through DR4 with a novel variant of rhTRAIL. J. Cell. Mol. Med. 2011, 15, 2216–2231. [Google Scholar] [CrossRef] [Green Version]
- Nolan, E.; Stikvoort, A.; Gurney, M.; Burduli, N.; Kirkham-McCarthy, L.; Daly, J.; Van De Donk, N.W.C.J.; Mutis, T.; Sarkar, S.; O’Dwyer, M.E. Targeting CD38high Acute Myeloid Leukaemia with “Affinity Optimized” Chimeric Antigen Receptor and Membrane Bound TRAIL Expressing Natural Killer Cells. Blood 2019, 134, 5536. [Google Scholar] [CrossRef]
- Van Dijk, M.; Murphy, E.; Morrell, R.; Knapper, S.; O’Dwyer, M.; Samali, A.; Szegezdi, E. The proteasome inhibitor bortezomib sensitizes AML with myelomonocytic differentiation to TRAIL mediated apoptosis. Cancers 2011, 3, 1329–1350. [Google Scholar] [CrossRef]
- Iyori, M.; Zhang, T.; Pantel, H.; Gagne, B.A.; Sentman, C.L. TRAIL/DR5 Plays a Critical Role in NK Cell-Mediated Negative Regulation of Dendritic Cell Cross-Priming of T Cells. J. Immunol. 2011, 187, 3087–3095. [Google Scholar] [CrossRef]
- Li, J.H.; Rosen, D.; Sondel, P.; Berke, G. Immune privilege and FasL: Two ways to inactivate effector cytotoxic T lymphocytes by FasL-expressing cells. Immunology 2002, 105, 267–277. [Google Scholar] [CrossRef] [PubMed]
- Romee, R.; Cooley, S.; Berrien-Elliott, M.M.; Westervelt, P.; Verneris, M.R.; Wagner, J.E.; Weisdorf, D.J.; Blazar, B.R.; Ustun, C.; DeFor, T.E.; et al. First-in-human phase 1 clinical study of the IL-15 superagonist complex ALT-803 to treat relapse after transplantation. Blood 2018, 131, 2515–2527. [Google Scholar] [CrossRef] [PubMed]
- Hills, R.K.; Castaigne, S.; Appelbaum, F.R.; Delaunay, J.; Petersdorf, S.; Othus, M.; Estey, E.H.; Dombret, H.; Chevret, S.; Ifrah, N.; et al. Addition of gemtuzumab ozogamicin to induction chemotherapy in adult patients with acute myeloid leukaemia: A meta-analysis of individual patient data from randomised controlled trials. Lancet Oncol. 2014, 15, 986–996. [Google Scholar] [CrossRef] [Green Version]
- Zheng, B.; Yu, S.F.; del Rosario, G.; Leong, S.R.; Lee, G.Y.; Vij, R.; Chiu, C.; Liang, W.C.; Wu, Y.; Chalouni, C.; et al. An anti–CLL-1 antibody–drug conjugate for the treatment of acute myeloid leukemia. Clin. Cancer Res. 2019, 25, 1358–1368. [Google Scholar] [CrossRef] [Green Version]
- Märklin, M.; Hagelstein, I.; Koerner, S.P.; Rothfelder, K.; Pfluegler, M.S.; Schumacher, A.; Grosse-Hovest, L.; Jung, G.; Salih, H.R. Bispecific NKG2D-CD3 and NKG2D-CD16 fusion proteins for induction of NK and T cell reactivity against acute myeloid leukemia. J. Immunother. Cancer 2019, 7. [Google Scholar] [CrossRef] [PubMed] [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] [Green Version]
- Arvindam, U.S.; van Hauten, P.M.M.; Schirm, D.; Schaap, N.; Hobo, W.; Blazar, B.R.; Vallera, D.A.; Dolstra, H.; Felices, M.; Miller, J.S. A trispecific killer engager molecule against CLEC12A effectively induces NK-cell mediated killing of AML cells. Leukemia 2020. [Google Scholar] [CrossRef] [PubMed]
- Chapuis, A.G.; Egan, D.N.; Bar, M.; Schmitt, T.M.; McAfee, M.S.; Paulson, K.G.; Voillet, V.; Gottardo, R.; Ragnarsson, G.B.; Bleakley, M.; et al. T cell receptor gene therapy targeting WT1 prevents acute myeloid leukemia relapse post-transplant. Nat. Med. 2019, 25, 1064–1072. [Google Scholar] [CrossRef] [PubMed]
- Kim, H.J.; Sohn, H.J.; Hong, J.A.; Lee, H.J.; Sohn, D.H.; Shin, C.A.; Il Cho, H.; Min, W.S.; Kim, T.G. Post-transplant immunotherapy with WT1-specific CTLs for high-risk acute myelogenous leukemia: A prospective clinical phase I/II trial. Bone Marrow Transplant. 2019, 54, 903–906. [Google Scholar] [CrossRef] [PubMed]
Autologous CAR-T | Allogeneic CAR-T | CAR-NK | |
---|---|---|---|
Efficacy | Established [11,12,15] | Investigational [22,23] | Investigational [17] |
Allogeneic Sources | NA | Yes | Yes |
Mechanism of Activation | CAR | CAR | CAR and Innate |
CRS/ICANS | Established | Anticipated | Likely Reduced [17] |
Cost of Product | $370,000–475,000 | Likely Reduced | Likely Reduced |
Cost of Care | Variable | Likely Equivalent | Potentially Reduced |
Cryopreservation | Established | Established | Investigational |
Viral Gene Delivery | Established, Feasible | Established, Feasible | Lower Efficiency |
Non-Viral (Stable) Gene Delivery | Clinical Trials [24,25] | Described in iPSCs [26] |
Target | Cases * | HSC * | LSC * | NK | CAR-NK Development |
---|---|---|---|---|---|
CD33 | 88% [141] | Yes | Yes | Yes | Clinical: CAR NK92 (NCT02944162) [140] Preclinical: Primary CD33 CAR-NK [142] |
CD123 | 78% [141] | Yes | Yes | No | Preclinical: Primary CAR-NK and CAR NK-92 [143,144,145] |
CLL-1 | 77% [146] | No | Yes | No | |
NKG2DL | 70% [87] | No | No | No | Clinical: AML (NCT04623944), CRC (NCT03415100) [130] Preclinical: Activity in Multiple Myeloma [147] |
CD38 | 70% [64] | No | No | Yes | Preclinical: PB CAR-NK and CAR-KHYG-1 [148] |
NPM1c | 35% [60] | No | Yes | No | Preclinical development of CIML-NK CAR [149] |
CD7 | 30% [150] | No | Yes | Yes | Preclinical: CD7 CAR-NK92 in T-ALL [151] |
TIM-3 | 87% [35] | No | Yes | Yes | |
CD96 | 51% [39] | No | Yes | Yes |
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 (http://creativecommons.org/licenses/by/4.0/).
Share and Cite
Gurney, M.; O’Dwyer, M. Realizing Innate Potential: CAR-NK Cell Therapies for Acute Myeloid Leukemia. Cancers 2021, 13, 1568. https://doi.org/10.3390/cancers13071568
Gurney M, O’Dwyer M. Realizing Innate Potential: CAR-NK Cell Therapies for Acute Myeloid Leukemia. Cancers. 2021; 13(7):1568. https://doi.org/10.3390/cancers13071568
Chicago/Turabian StyleGurney, Mark, and Michael O’Dwyer. 2021. "Realizing Innate Potential: CAR-NK Cell Therapies for Acute Myeloid Leukemia" Cancers 13, no. 7: 1568. https://doi.org/10.3390/cancers13071568
APA StyleGurney, M., & O’Dwyer, M. (2021). Realizing Innate Potential: CAR-NK Cell Therapies for Acute Myeloid Leukemia. Cancers, 13(7), 1568. https://doi.org/10.3390/cancers13071568