Redirecting the Immune Microenvironment in Acute Myeloid Leukemia
Abstract
:Simple Summary
Abstract
1. Introduction
2. Acute Myeloid Leukemia Harnesses the Immunological Microenvironment
2.1. Human Leukocyte Antigens
2.2. Checkpoint Molecules
2.3. T-cellular Immune Dysregulation
2.4. NK Cell-Related Strategies of Immune Evasion
3. Therapeutic Approaches Redirecting Immuno-Suppressive Microenvironment
3.1. Checkpoint Inhibitors
3.2. Antibody-Based Therapy
3.3. Cell-Based Therapy
3.4. Therapeutic Impact of Cytokines and Immune-Modulating Factors
3.5. Vaccines
3.6. Oncolytic Viruses
4. Conclusions
Supplementary Materials
Author Contributions
Funding
Acknowledgments
Conflicts of Interest
References
- Ladikou, E.E.; Sivaloganathan, H.; Pepper, A.; Chevassut, T. Acute Myeloid Leukaemia in Its Niche: The Bone Marrow Microenvironment in Acute Myeloid Leukaemia. Curr. Oncol. Rep. 2020, 22. [Google Scholar] [CrossRef] [Green Version]
- Rasche, M.; Zimmermann, M.; Borschel, L.; Bourquin, J.; Dworzak, M.; Klingebiel, T.; Lehrnbecher, T.; Creutzig, U.; Klusmann, J.; Reinhardt, D. Successes and challenges in the treatment of pediatric acute myeloid leukemia: A retrospective analysis of the AML-BFM trials from 1987 to 2012. Leukemia 2018, 32, 2167–2177. [Google Scholar] [CrossRef] [Green Version]
- Rasche, M.; Steidel, E.; Kondryn, D.; Von Neuhoff, N.; Sramkova, L.; Creutzig, U.; Dworzak, M.; Reinhardt, D. Impact of a Risk-Adapted Treatment Approach in Pediatric AML: A Report of the AML-BFM Registry 2012. Blood 2019, 134, 293. [Google Scholar] [CrossRef]
- Li, S.; Mason, C.E.; Melnick, A. Genetic and epigenetic heterogeneity in acute myeloid leukemia. Curr. Opin. Genet. Dev. 2016, 36, 100–106. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Davila, J.; Slotkin, E.; Renaud, T. Relapsed and refractory pediatric acute myeloid leukemia: Current and emerging treatments. Paediatr. Drugs 2014, 16, 151–168. [Google Scholar] [CrossRef] [PubMed]
- Winer, E.S.; Stone, R.M. Novel therapy in Acute myeloid leukemia (AML): Moving toward targeted approaches. Ther. Adv. Hematol. 2019, 10. [Google Scholar] [CrossRef]
- Petersdorf, S.H.; Kopecky, K.J.; Slovak, M.; Willman, C.; Nevill, T.; Brandwein, J.; Larson, R.A.; Erba, H.P.; Stiff, P.J.; Stuart, R.K.; et al. A phase 3 study of gemtuzumab ozogamicin during induction and postconsolidation therapy in younger patients with acute myeloid leukemia. Blood 2013, 4854–4860. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- He, S.Z.; Busfield, S.; Ritchie, D.S.; Hertzberg, M.S.; Durrant, S.; Lewis, I.D.; Marlton, P.; McLachlan, A.J.; Kerridge, I.; Bradstock, K.F.; et al. A Phase 1 study of the safety, pharmacokinetics and anti-leukemic activity of the anti-CD123 monoclonal antibody CSL360 in relapsed, refractory or high-risk acute myeloid leukemia. Leuk. Lymphoma 2015, 56. [Google Scholar] [CrossRef]
- Feldman, E.J.; Brandwein, J.; Stone, R.; Kalaycio, M.; Moore, J.; O’Connor, J.; Wedel, N.; Roboz, G.J.; Miller, C.; Chopra, R.; et al. Phase III randomized multicenter study of a humanized anti-CD33 monoclonal antibody, lintuzumab, in combination with chemotherapy, versus chemotherapy alone in patients with refractory or first-relapsed acute myeloid leukemia. J. Clin. Oncol. 2005, 23. [Google Scholar] [CrossRef] [PubMed]
- Morsink, L.M.; Walter, R.B. Novel monoclonal antibody-based therapies for acute myeloid leukemia. Best Pract. Res. Clin. Haematol. 2019, 32, 116–126. [Google Scholar] [CrossRef]
- Albring, J.C.; Inselmann, S.; Sauer, T.; Schliemann, C.; Altvater, B.; Kailayangiri, S.; Rössig, C.; Hartmann, W.; Knorrenschild, J.R.; Sohlbach, K.; et al. PD-1 checkpoint blockade in patients with relapsed AML after allogeneic stem cell transplantation. Bone Marrow Transpl. 2017, 317–320. [Google Scholar] [CrossRef]
- Bashey, A.; Medina, B.; Corringham, S.; Pasek, M.; Carrier, E.; Vrooman, L.; Lowy, I.; Solomon, S.R.; Morris, L.E.; Holland, H.K.; et al. CTLA4 blockade with ipilimumab to treat relapse of malignancy after allogeneic hematopoietic cell transplantation. Blood 2009, 113, 1581–1588. [Google Scholar] [CrossRef] [PubMed]
- Davids, M.S.; Kim, H.T.; Bachireddy, P.; Costello, C.; Liguori, R.; Savell, A.; Lukez, A.P.; Avigan, D.; Chen, Y.-B.; McSweeney, P.; et al. Ipilimumab for Patients with Relapse after Allogeneic Transplantation. N. Engl. J. Med. 2016, 375, 143–153. [Google Scholar] [CrossRef] [PubMed]
- Kruger, S.; Ilmer, M.; Kobold, S.; Cadilha, B.L.; Endres, S.; Ormanns, S.; Schuebbe, G.; Renz, B.W.; D’Haese, J.G.; Schloesser, H.; et al. Advances in cancer immunotherapy 2019—latest trends. J. Exp. Clin. Cancer Res. 2019, 38, 1–11. [Google Scholar] [CrossRef] [Green Version]
- Sweeney, C.; Vyas, P. The Graft-Versus-Leukemia Effect in AML. Front. Oncol. 2019, 9. [Google Scholar] [CrossRef]
- Vago, L.; Gojo, I. Immune escape and immunotherapy of acute myeloid leukemia. J. Clin. Investig. 2020, 130, 1552–1564. [Google Scholar] [CrossRef]
- Stölzel, F.; Hackmann, K.; Kuithan, F.; Mohr, B.; Füssel, M.; Oelschlägel, U.; Thiede, C.; Röllig, C.; Platzbecker, U.; Schetelig, J.; et al. Clonal evolution including partial loss of human leukocyte antigen genes favoring extramedullary acute myeloid leukemia relapse after matched related allogeneic hematopoietic stem cell transplantation. Transplantation 2012, 93, 744–749. [Google Scholar] [CrossRef]
- Toffalori, C.; Riba, M.; Zito, L.; Barcella, M.; Spinelli, O.; Crucitti, L.; Cieri, N.; Peccatori, J.; Bernardi, M.; Bonini, C.; et al. Acute Myeloid Leukemia Relapses after Allogenenic HSCT Display a Distinctive Immune-Related Signature, with Frequent and Functionally Relevant Alterations in HLA Class II Antigen Presentation and T Cell Costimulation. Blood 2014, 124, 427. [Google Scholar] [CrossRef]
- Crucitti, L.; Crocchiolo, R.; Toffalori, C.; Mazzi, B.; Greco, R.; Signori, A.; Sizzano, F.; Chiesa, L.; Zino, E.; Lupo Stanghellini, M.T.; et al. Incidence, risk factors and clinical outcome of leukemia relapses with loss of the mismatched HLA after partially incompatible hematopoietic stem cell transplantation. Leukemia 2015, 29, 1143–1152. [Google Scholar] [CrossRef] [PubMed]
- Christopher, M.J.; Petti, A.A.; Rettig, M.P.; Miller, C.A.; Chendamarai, E.; Duncavage, E.J.; Klco, J.M.; Helton, N.M.; O’Laughlin, M.; Fronick, C.C.; et al. Immune Escape of Relapsed AML Cells after Allogeneic Transplantation. N. Engl. J. Med. 2018, 379, 2330–2341. [Google Scholar] [CrossRef]
- Colonna, M.; Samaridis, J.; Cella, M.; Angman, L.; Allen, R.L.; O’Callaghan, C.A.; Dunbar, R.; Ogg, G.S.; Cerundolo, V.; Rolink, A. Human myelomonocytic cells express an inhibitory receptor for classical and nonclassical MHC class I molecules. J. Immunol. 1998, 160, 3096–3100. [Google Scholar]
- Lin, A.; Yan, W.-H. Human Leukocyte Antigen-G (HLA-G) Expression in Cancers: Roles in Immune Evasion, Metastasis and Target for Therapy. Mol. Med. 2015, 21, 782–791. [Google Scholar] [CrossRef]
- Gros, F.; Sebti, Y.; de Guibert, S.; Branger, B.; Bernard, M.; Fauchet, R.; Amiot, L. Soluble HLA-G molecules increase during acute leukemia, especially in subtypes affecting monocytic and lymphoid lineages. Neoplasia 2006, 8, 223–230. [Google Scholar] [CrossRef] [PubMed]
- Mizuno, S.; Emi, N.; Kasai, M.; Ishitani, A.; Saito, H. Aberrant expression of HLA-G antigen in interferon gamma-stimulated acute myelogenous leukaemia. Br. J. Haematol. 2000, 111. [Google Scholar] [CrossRef] [PubMed]
- Yan, W.-H.; Lin, A.; Chen, B.-G.; Luo, W.-D.; Dai, M.-Z.; Chen, X.-J.; Xu, H.-H.; Li, B.-L. Unfavourable clinical implications for HLA-G expression in acute myeloid leukaemia. J. Cell. Mol. Med. 2008, 12, 889–898. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Hamed, N.M.; El Halawani, N.; Nafea, D.; El Rahman, M.; Kasber, A. Soluble HLA-G: A novel marker in acute myeloid leukemia patients. Acta Med. Int. 2017, 4, 51. [Google Scholar] [CrossRef]
- Guo, Q.Y.; Chen, B.G.; Ruan, Y.Y.; Lin, A.; Yan, W.H. HLA-G expression is irrelevant to prognosis in patients with acute myeloid leukemia. Leuk. Res. 2011, 35, 1350–1354. [Google Scholar] [CrossRef] [PubMed]
- Poláková, K.; Krcová, M.; Kuba, D.; Russ, G. Analysis of HLA-G expression in malignant hematopoetic cells from leukemia patients. Leuk. Res. 2003, 27. [Google Scholar] [CrossRef]
- Zhang, L.; Gajewski, T.F.; Kline, J. PD-1/PD-L1 interactions inhibit antitumor immune responses in a murine acute myeloid leukemia model. Blood 2009, 114, 1545–1552. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Zhou, Q.; Munger, M.E.; Highfill, S.L.; Tolar, J.; Weigel, B.J.; Riddle, M.; Sharpe, A.H.; Vallera, D.A.; Azuma, M.; Levine, B.L.; et al. Program death-1 signaling and regulatory T cells collaborate to resist the function of adoptively transferred cytotoxic T lymphocytes in advanced acute myeloid leukemia. Blood 2010, 116, 2484–2493. [Google Scholar] [CrossRef] [PubMed]
- Zhou, Q.; Munger, M.E.; Veenstra, R.G.; Weigel, B.J.; Hirashima, M.; Munn, D.H.; Murphy, W.J.; Azuma, M.; Anderson, A.C.; Kuchroo, V.K.; et al. Coexpression of Tim-3 and PD-1 identifies a CD8+ T-cell exhaustion phenotype in mice with disseminated acute myelogenous leukemia. Blood 2011, 117, 4501–4510. [Google Scholar] [CrossRef] [PubMed]
- Kikushige, Y.; Miyamoto, T.; Yuda, J.; Jabbarzadeh-Tabrizi, S.; Shima, T.; Takayanagi, S.-i.; Niiro, H.; Yurino, A.; Miyawaki, K.; Takenaka, K.; et al. A TIM-3/Gal-9 Autocrine Stimulatory Loop Drives Self-Renewal of Human Myeloid Leukemia Stem Cells and Leukemic Progression. Cell Stem Cell 2015, 17, 341–352. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Gonçalves, S.I.; Yasinska, I.M.; Sakhnevych, S.S.; Fiedler, W.; Wellbrock, J.; Bardelli, M.; Varani, L.; Hussain, R.; Siligardi, G.; Ceccone, G.; et al. The Tim-3-galectin-9 Secretory Pathway is Involved in the Immune Escape of Human Acute Myeloid Leukemia Cells. EBioMedicine 2017, 22. [Google Scholar] [CrossRef] [Green Version]
- Chen, C.; Liang, C.; Wang, S.; Chio, C.L.; Zhang, Y.; Zeng, C.; Chen, S.; Wang, C.; Li, Y. Expression patterns of immune checkpoints in acute myeloid leukemia. J. Hematol. Oncol. 2020, 13, 28. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Knaus, H.A.; Berglund, S.; Hackl, H.; Blackford, A.L.; Zeidner, J.F.; Montiel-Esparza, R.; Mukhopadhyay, R.; Vanura, K.; Blazar, B.R.; Karp, J.E.; et al. Signatures of CD8+ T cell dysfunction in AML patients and their reversibility with response to chemotherapy. JCI Insight 2018, 3. [Google Scholar] [CrossRef] [Green Version]
- Li, Z.; Philip, M.; Ferrell, P.B. Alterations of T-cell-mediated immunity in acute myeloid leukemia. Oncogene 2020, 39, 3611–3619. [Google Scholar] [CrossRef]
- Mussai, F.; De Santo, C.; Abu-Dayyeh, I.; Booth, S.; Quek, L.; McEwen-Smith, R.M.; Qureshi, A.; Dazzi, F.; Vyas, P.; Cerundolo, V. Acute myeloid leukemia creates an arginase-dependent immunosuppressive microenvironment. Blood 2013, 122, 749–758. [Google Scholar] [CrossRef] [Green Version]
- Wang, X.; Zheng, J.; Liu, J.; Yao, J.; He, Y.; Li, X.; Yu, J.; Yang, J.; Liu, Z.; Huang, S. Increased population of CD4(+)CD25(high), regulatory T cells with their higher apoptotic and proliferating status in peripheral blood of acute myeloid leukemia patients. Eur. J. Haematol. 2005, 75, 468–476. [Google Scholar] [CrossRef]
- Szczepanski, M.J.; Szajnik, M.; Czystowska, M.; Mandapathil, M.; Strauss, L.; Welsh, A.; Foon, K.A.; Whiteside, T.L.; Boyiadzis, M. Increased frequency and suppression by regulatory T cells in patients with acute myelogenous leukemia. Clin. Cancer Res. 2009, 15, 3325–3332. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Liao, W.; Lin, J.-X.; Leonard, W.J. Interleukin-2 at the crossroads of effector responses, tolerance, and immunotherapy. Immunity 2013, 38, 13–25. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Ettinghausen, S.E.; Lipford, E.H.; Mulé, J.J.; Rosenberg, S.A. Recombinant interleukin 2 stimulates in vivo proliferation of adoptively transferred lymphokine-activated killer (LAK) cells. J. Immunol. 1985, 135, 3623. [Google Scholar]
- Arandi, N.; Ramzi, M.; Safaei, F.; Monabati, A. Overexpression of indoleamine 2,3-dioxygenase correlates with regulatory T cell phenotype in acute myeloid leukemia patients with normal karyotype. Blood Res. 2018, 53, 294–298. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Curti, A.; Pandolfi, S.; Valzasina, B.; Aluigi, M.; Isidori, A.; Ferri, E.; Salvestrini, V.; Bonanno, G.; Rutella, S.; Durelli, I.; et al. Modulation of tryptophan catabolism by human leukemic cells results in the conversion of CD25- into CD25+ T regulatory cells. Blood 2007, 109, 2871–2877. [Google Scholar] [CrossRef] [PubMed]
- Han, Y.; Dong, Y.; Yang, Q.; Xu, W.; Jiang, S.; Yu, Z.; Yu, K.; Zhang, S. Acute Myeloid Leukemia Cells Express ICOS Ligand to Promote the Expansion of Regulatory T Cells. Front. Immunol. 2018, 9, 2227. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Chen, W.; Jin, W.; Hardegen, N.; Lei, K.J.; Li, L.; Marinos, N.; McGrady, G.; Wahl, S.M. Conversion of peripheral CD4+CD25- naive T cells to CD4+CD25+ regulatory T cells by TGF-beta induction of transcription factor Foxp3. J. Exp. Med. 2003, 198. [Google Scholar] [CrossRef]
- Buggins, A.G.; Milojkovic, D.; Arno, M.J.; Lea, N.C.; Mufti, G.J.; Thomas, N.S.; Hirst, W.J. Microenvironment produced by acute myeloid leukemia cells prevents T cell activation and proliferation by inhibition of NF-kappaB, c-Myc, and pRb pathways. J. Immunol. 2001, 167, 6021–6030. [Google Scholar] [CrossRef] [Green Version]
- Orleans-Lindsay, J.K.; Barber, L.D.; Prentice, H.G.; Lowdell, M.W. Acute myeloid leukaemia cells secrete a soluble factor that inhibits T and NK cell proliferation but not cytolytic function--implications for the adoptive immunotherapy of leukaemia. Clin. Exp. Immunol. 2001, 126, 403–411. [Google Scholar] [CrossRef]
- Schnorfeil, F.M.; Lichtenegger, F.S.; Emmerig, K.; Schlueter, M.; Neitz, J.S.; Draenert, R.; Hiddemann, W.; Subklewe, M. T cells are functionally not impaired in AML: Increased PD-1 expression is only seen at time of relapse and correlates with a shift towards the memory T cell compartment. J. Hematol. Oncol. 2015, 8. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Hasegawa, K.; Tanaka, S.; Fujiki, F.; Morimoto, S.; Nakajima, H.; Tatsumi, N.; Nakata, J.; Takashima, S.; Nishida, S.; Tsuboi, A.; et al. An Immunocompetent Mouse Model for MLL/AF9 Leukemia Reveals the Potential of Spontaneous Cytotoxic T-Cell Response to an Antigen Expressed in Leukemia Cells. PLoS ONE 2015, 10, e0144594. [Google Scholar] [CrossRef]
- Le Dieu, R.; Taussig, D.C.; Ramsay, A.G.; Mitter, R.; Miraki-Moud, F.; Fatah, R.; Lee, A.M.; Lister, T.A.; Gribben, J.G. Peripheral blood T cells in acute myeloid leukemia (AML) patients at diagnosis have abnormal phenotype and genotype and form defective immune synapses with AML blasts. Blood 2009, 114, 3909–3916. [Google Scholar] [CrossRef] [Green Version]
- Jia, B.; Zhao, C.; Rakszawski, K.L.; Claxton, D.F.; Ehmann, W.C.; Rybka, W.B.; Mineishi, S.; Wang, M.; Shike, H.; Bayerl, M.G.; et al. Eomes+T-betlow CD8+ T Cells Are Functionally Impaired and Are Associated with Poor Clinical Outcome in Patients with Acute Myeloid Leukemia. Cancer Res. 2019, 79, 1635–1645. [Google Scholar] [CrossRef]
- van Galen, P.; Hovestadt, V.; Wadsworth Ii, M.H.; Hughes, T.K.; Griffin, G.K.; Battaglia, S.; Verga, J.A.; Stephansky, J.; Pastika, T.J.; Lombardi Story, J.; et al. Single-Cell RNA-Seq Reveals AML Hierarchies Relevant to Disease Progression and Immunity. Cell 1265, 76, 1265–1281.e24. [Google Scholar] [CrossRef] [Green Version]
- 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-beta1. Haematologica 2011, 96, 1302–1309. [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 NCRdull phenotype induction. Blood 2007, 109, 323–330. [Google Scholar] [CrossRef] [PubMed]
- Costello, R.T.; Sivori, S.; Marcenaro, E.; Lafage-Pochitaloff, M.; Mozziconacci, M.-J.; Reviron, D.; Gastaut, J.-A.; Pende, D.; Olive, D.; Moretta, A. Defective expression and function of natural killer cell-triggering receptors in patients with acute myeloid leukemia. Blood 2002, 99, 3661–3667. [Google Scholar] [CrossRef] [PubMed]
- Venton, G.; Labiad, Y.; Colle, J.; Fino, A.; Afridi, S.; Torres, M.; Monteuil, S.; Loriod, B.; Fernandez-Nunez, N.; Farnault, L.; et al. Natural killer cells in acute myeloid leukemia patients: From phenotype to transcriptomic analysis. Immunol. Res. 2016, 64, 1225–1236. [Google Scholar] [CrossRef]
- Nowbakht, P.; Ionescu, M.-C.S.; Rohner, A.; Kalberer, C.P.; Rossy, E.; Mori, L.; Cosman, D.; De Libero, G.; Wodnar-Filipowicz, A. Ligands for natural killer cell-activating receptors are expressed upon the maturation of normal myelomonocytic cells but at low levels in acute myeloid leukemias. Blood 2005, 105, 3615–3622. [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] [PubMed] [Green Version]
- Mundy-Bosse, B.; Kathleen, M.; Mao, C.; Ahmed, E.; Chen, L.; Scoville, S.D.; Freud, A.G.; Yu, J.; Caligiuri, M.A. Acute Myeloid Leukemia Alters Natural Killer Cell Maturation and Functional Activation. Blood 2014, 124, 754. [Google Scholar] [CrossRef]
- Hyun, S.Y.; Na, E.J.; Jang, J.E.; Chung, H.; Kim, S.J.; Kim, J.S.; Kong, J.H.; Shim, K.Y.; Lee, J.I.; Min, Y.H.; et al. Immunosuppressive role of CD11b+ CD33+ HLA-DR- myeloid-derived suppressor cells-like blast subpopulation in acute myeloid leukemia. Cancer Med. 2020. [Google Scholar] [CrossRef] [PubMed]
- Tumino, N.; Besi, F.; Di Pace, A.L.; Mariotti, F.R.; Merli, P.; Li Pira, G.; Galaverna, F.; Pitisci, A.; Ingegnere, T.; Pelosi, A.; et al. PMN-MDSC are a new target to rescue graft-versus-leukemia activity of NK cells in haplo-HSC transplantation. Leukemia 2020, 34, 932–937. [Google Scholar] [CrossRef]
- Sobash, P.T.; Kolhe, R.; Karim, N.A.; Guddati, A.K.; Jillella, A.; Kota, V. Role of indoleamine 2,3-dioxygenase in acute myeloid leukemia. Future Oncol. 2020. [Google Scholar] [CrossRef]
- Gao, L.; Yu, S.; Zhang, X. Hypothesis: Tim-3/galectin-9, a new pathway for leukemia stem cells survival by promoting expansion of myeloid-derived suppressor cells and differentiating into tumor-associated macrophages. Cell Biochem. Biophys. 2014, 70, 273–277. [Google Scholar] [CrossRef] [PubMed]
- Pyzer, A.R.; Stroopinsky, D.; Rajabi, H.; Washington, A.; Tagde, A.; Coll, M.; Fung, J.; Bryant, M.P.; Cole, L.; Palmer, K.; et al. MUC1-mediated induction of myeloid-derived suppressor cells in patients with acute myeloid leukemia. Blood 2017, 129, 1791–1801. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- López-Larrea, C.; Suárez-Alvarez, B.; López-Soto, A.; López-Vázquez, A.; Gonzalez, S. The NKG2D receptor: Sensing stressed cells. Trends Mol. Med. 2008, 14. [Google Scholar] [CrossRef] [PubMed]
- Pende, D.; Spaggiari, G.M.; Marcenaro, S.; Martini, S.; Rivera, P.; Capobianco, A.; Falco, M.; Lanino, E.; Pierri, I.; Zambello, R.; et al. Analysis of the receptor-ligand interactions in the natural killer-mediated lysis of freshly isolated myeloid or lymphoblastic leukemias: Evidence for the involvement of the Poliovirus receptor (CD155) and Nectin-2 (CD112). Blood 2005, 105, 2066–2073. [Google Scholar] [CrossRef] [PubMed]
- Hong, C.-S.; Muller, L.; Whiteside, T.L.; Boyiadzis, M. Plasma exosomes as markers of therapeutic response in patients with acute myeloid leukemia. Front. Immunol. 2014, 5, 160. [Google Scholar] [CrossRef] [Green Version]
- 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]
- 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]
- 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]
- Spaggiari, G.M.; Capobianco, A.; Abdelrazik, H.; Becchetti, F.; Mingari, M.C.; Moretta, L. Mesenchymal stem cells inhibit natural killer-cell proliferation, cytotoxicity, and cytokine production: Role of indoleamine 2,3-dioxygenase and prostaglandin E2. Blood 2008, 111. [Google Scholar] [CrossRef]
- Joshi, P.C.; Zhou, X.; Cuchens, M.; Jones, Q. Prostaglandin E2 suppressed IL-15-mediated human NK cell function through down-regulation of common gamma-chain. J. Immunol. 2001, 166, 885–891. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Vacchelli, E.; Aranda, F.; Eggermont, A.; Sautès-Fridman, C.; Tartour, E.; Kennedy, E.P.; Platten, M.; Zitvogel, L.; Kroemer, G.; Galluzzi, L. Trial watch: IDO inhibitors in cancer therapy. Oncoimmunology 2014, 3, e957994. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Della Chiesa, M.; Carlomagno, S.; Frumento, G.; Balsamo, M.; Cantoni, C.; Conte, R.; Moretta, L.; Moretta, A.; Vitale, M. The tryptophan catabolite L-kynurenine inhibits the surface expression of NKp46- and NKG2D-activating receptors and regulates NK cell function. Blood 2006, 108, 4118–4125. [Google Scholar] [CrossRef]
- Mansour, I.; Zayed, R.A.; Said, F.; Latif, L.A. Indoleamine 2,3-dioxygenase and regulatory T cells in acute myeloid leukemia. Hematology 2016, 21, 447–453. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Sharma, M.D.; Baban, B.; Chandler, P.; Hou, D.-Y.; Singh, N.; Yagita, H.; Azuma, M.; Blazar, B.R.; Mellor, A.L.; Munn, D.H. Plasmacytoid dendritic cells from mouse tumor-draining lymph nodes directly activate mature Tregs via indoleamine 2,3-dioxygenase. J. Clin. Investig. 2007, 117, 2570–2582. [Google Scholar] [CrossRef] [Green Version]
- Munn, D.H.; Mellor, A.L. IDO in the Tumor Microenvironment: Inflammation, Counter-Regulation, and Tolerance. Trends Immunol. 2016, 37, 193–207. [Google Scholar] [CrossRef] [Green Version]
- 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-beta-dependent manner. J. Exp. Med. 2005, 202, 1075–1085. [Google Scholar] [CrossRef]
- Hargadon, K.M.; Johnson, C.E.; Williams, C.J. Immune checkpoint blockade therapy for cancer: An overview of FDA-approved immune checkpoint inhibitors. Int. Immunopharmacol. 2018, 62, 29–39. [Google Scholar] [CrossRef]
- Liao, D.; Wang, M.; Liao, Y.; Li, J.; Niu, T. A Review of Efficacy and Safety of Checkpoint Inhibitor for the Treatment of Acute Myeloid Leukemia. Front. Pharmacol. 2019, 10. [Google Scholar] [CrossRef] [Green Version]
- Davids, M.S.; Kim, H.T.; Costello, C.; Herrera, A.F.; Locke, F.L.; Maegawa, R.O.; Savell, A.; Mazzeo, M.; Anderson, A.; Boardman, A.P.; et al. A multicenter phase 1 study of nivolumab for relapsed hematologic malignancies after allogeneic transplantation. Blood 2020, 135, 2182–2191. [Google Scholar] [CrossRef]
- Shallis, R.M.; Bewersdorf, J.P.; Gowda, L.; Podoltsev, N.A.; Prebet, T.; Gore, S.D.; Halene, S.; Isufi, I.; Foss, F.M.; Huntington, S.F.; et al. Immune Checkpoint Inhibitor Therapy for Acute Myeloid Leukemia and Higher-Risk Myelodysplastic Syndromes: A Single-Center Experience. Blood 2019, 134, 1330. [Google Scholar] [CrossRef]
- 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]
- Assi, R.; Kantarjian, H.M.; Daver, N.G.; Garcia-Manero, G.; Benton, C.B.; Thompson, P.A.; Borthakur, G.; Kadia, T.M.; Alvarado, Y.; Jabbour, E.J.; et al. Results of a Phase 2, Open-Label Study of Idarubicin (I), Cytarabine (A) and Nivolumab (Nivo) in Patients with Newly Diagnosed Acute Myeloid Leukemia (AML) and High-Risk Myelodysplastic Syndrome (MDS). Blood 2018, 132, 905. [Google Scholar] [CrossRef]
- Daver, N.; Basu, S.; Garcia-Manero, G.; Cortes, J.E.; Ravandi, F.; Jabbour, E.J.; Hendrickson, S.; Pierce, S.; Ning, J.; Konopleva, M.; et al. Phase IB/II Study of Nivolumab in Combination with Azacytidine (AZA) in Patients (pts) with Relapsed Acute Myeloid Leukemia (AML). Blood 2016, 128, 763. [Google Scholar] [CrossRef]
- Ghosh, A.; Barba, P.; Perales, M.-A. Checkpoint inhibitors in AML: Are we there yet? Br. J. Haematol. 2020, 188, 159–167. [Google Scholar] [CrossRef] [PubMed]
- Karim, L.A.; Wang, P.; de Guzman, J.; Higgins, B.; Chahine, J.; Sheehan, C.; Kallakury, B.; Ross, J.S. Abstract 3724: PDL1 protein expression and tumor mutation burden in hematologic malignancies: Correlation with Hodgkin and high grade lymphoma. In Clinical Research (Excluding Clinical Trials); American Association for Cancer Research: Washington, DC, USA, 2017; p. 3724. [Google Scholar]
- Yang, H.; Bueso-Ramos, C.; DiNardo, C.; Estecio, M.R.; Davanlou, M.; Geng, Q.-R.; Fang, Z.; Nguyen, M.; Pierce, S.; Wei, Y.; et al. Expression of PD-L1, PD-L2, PD-1 and CTLA4 in myelodysplastic syndromes is enhanced by treatment with hypomethylating agents. Leukemia 2014, 28, 1280–1288. [Google Scholar] [CrossRef]
- Lindblad, K.E.; Goswami, M.; Hourigan, C.S.; Oetjen, K.A. Immunological effects of hypomethylating agents. Expert Rev. Hematol. 2017, 10, 745–752. [Google Scholar] [CrossRef] [PubMed]
- Kikushige, Y.; Shima, T.; Takayanagi, S.-i.; Urata, S.; Miyamoto, T.; Iwasaki, H.; Takenaka, K.; Teshima, T.; Tanaka, T.; Inagaki, Y.; et al. TIM-3 is a promising target to selectively kill acute myeloid leukemia stem cells. Cell Stem Cell 2010, 7, 708–717. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Dama, P.; Tang, M.; Fulton, N.; Kline, J.; Liu, H. Gal9/Tim-3 expression level is higher in AML patients who fail chemotherapy. J. Immunother. Cancer 2019, 7, 175. [Google Scholar] [CrossRef]
- Williams, P.; Basu, S.; Garcia-Manero, G.; Hourigan, C.S.; Oetjen, K.A.; Cortes, J.E.; Ravandi, F.; Jabbour, E.J.; Al-Hamal, Z.; Konopleva, M.; et al. The distribution of T-cell subsets and the expression of immune checkpoint receptors and ligands in patients with newly diagnosed and relapsed acute myeloid leukemia. Cancer 2019, 125, 1470–1481. [Google Scholar] [CrossRef] [Green Version]
- Broglie, L.; Gershan, J.; Burke, M.J. Checkpoint inhibition of PD-L1 and CTLA-4 in a child with refractory acute leukemia. Int. J. Hematol. Oncol. 2019, 8, IJH10. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Dutta, S.; Saxena, R. The Expression Pattern of CD33 Antigen Can Differentiate Leukemic from Normal Progenitor Cells in Acute Myeloid Leukemia. Indian J. Hematol. Blood Transfus. 2014, 30, 130–134. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Pelosi, E.; Castelli, G.; Testa, U. Targeting LSCs through membrane antigens selectively or preferentially expressed on these cells. Blood Cells Mol. Dis. 2015, 55, 336–346. [Google Scholar] [CrossRef]
- Creutzig, U.; Harbott, J.; Sperling, C.; Ritter, J.; Zimmermann, M.; Loffler, H.; Riehm, H.; Schellong, G.; Ludwig, W.D. Clinical significance of surface antigen expression in children with acute myeloid leukemia: Results of study AML-BFM-87. Blood 1995, 86, 3097–3108. [Google Scholar] [CrossRef] [Green Version]
- Bras, A.E.; De Haas, V.; van Stigt, A.; Jongen-Lavrencic, M.; Beverloo, H.B.; Te Marvelde, J.G.; Zwaan, C.M.; van Dongen, J.J.M.; Leusen, J.H.W.; van der Velden, V.H.J. CD123 expression levels in 846 acute leukemia patients based on standardized immunophenotyping. Cytom. B Clin. Cytom. 2019, 96, 134–142. [Google Scholar] [CrossRef]
- Zwaan, C.M.; Reinhardt, D.; Corbacioglu, S.; van Wering, E.R.; Bökkerink, J.P.M.; Tissing, W.J.E.; Samuelsson, U.; Feingold, J.; Creutzig, U.; Kaspers, G.J.L. Gemtuzumab ozogamicin: First clinical experiences in children with relapsed/refractory acute myeloid leukemia treated on compassionate-use basis. Blood 2003, 101, 3868–3871. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Niktoreh, N.; Lerius, B.; Zimmermann, M.; Gruhn, B.; Escherich, G.; Bourquin, J.P.; Dworzak, M.; Sramkova, L.; Rossig, C.; Creutzig, U.; et al. Gemtuzumab ozogamicin in children with relapsed or refractory acute myeloid leukemia: A report by Berlin-Frankfurt-Münster study group. Haematologica 2019, 104. [Google Scholar] [CrossRef] [Green Version]
- Renneville, A.; Abdelali, R.B.; Chevret, S.; Nibourel, O.; Cheok, M.; Pautas, C.; Duléry, R.; Boyer, T.; Cayuela, J.-M.; Hayette, S.; et al. Clinical impact of gene mutations and lesions detected by SNP-array karyotyping in acute myeloid leukemia patients in the context of gemtuzumab ozogamicin treatment: Results of the ALFA-0701 trial. Oncotarget 2014, 5, 916–932. [Google Scholar] [CrossRef] [Green Version]
- Gamis, A.S.; Alonzo, T.A.; Meshinchi, S.; Sung, L.; Gerbing, R.B.; Raimondi, S.C.; Hirsch, B.A.; Kahwash, S.B.; Heerema-McKenney, A.; Winter, L.; et al. Gemtuzumab ozogamicin in children and adolescents with de novo acute myeloid leukemia improves event-free survival by reducing relapse risk: Results from the randomized phase III Children’s Oncology Group trial AAML0531. J. Clin. Oncol. 2014, 32, 3021–3032. [Google Scholar] [CrossRef] [Green Version]
- Daver, N.G.; Montesinos, P.; DeAngelo, D.J.; Wang, E.S.; Papadantonakis, N.; Deconinck, E.; Erba, H.P.; Pemmaraju, N.; Lane, A.A.; Rizzieri, D.A.; et al. Clinical Profile of IMGN632, a Novel CD123-Targeting Antibody-Drug Conjugate (ADC), in Patients with Relapsed/Refractory (R/R) Acute Myeloid Leukemia (AML) or Blastic Plasmacytoid Dendritic Cell Neoplasm (BPDCN). Blood 2019, 134, 734. [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] [PubMed] [Green Version]
- Bakker, A.B.H.; van den Oudenrijn, S.; Bakker, A.Q.; Feller, N.; van Meijer, M.; Bia, J.A.; Jongeneelen, M.A.C.; Visser, T.J.; Bijl, N.; Geuijen, C.A.W.; et al. C-type lectin-like molecule-1: A novel myeloid cell surface marker associated with acute myeloid leukemia. Cancer Res. 2004, 64, 8443–8450. [Google Scholar] [CrossRef] [Green Version]
- van Rhenen, A.; van Dongen, G.A.M.S.; Kelder, A.; Rombouts, E.J.; Feller, N.; Moshaver, B.; Stigter-van Walsum, M.; Zweegman, S.; Ossenkoppele, G.J.; Jan Schuurhuis, G. The novel AML stem cell associated antigen CLL-1 aids in discrimination between normal and leukemic stem cells. Blood 2007, 110, 2659–2666. [Google Scholar] [CrossRef] [Green Version]
- Jiang, Y.-P.; Liu, B.Y.; Zheng, Q.; Panuganti, S.; Chen, R.; Zhu, J.; Mishra, M.; Huang, J.; Dao-Pick, T.; Roy, S.; et al. CLT030, a leukemic stem cell-targeting CLL1 antibody-drug conjugate for treatment of acute myeloid leukemia. Blood Adv. 2018, 2, 1738–1749. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Lu, H.; Zhou, Q.; Deshmukh, V.; Phull, H.; Ma, J.; Tardif, V.; Naik, R.R.; Bouvard, C.; Zhang, Y.; Choi, S.; et al. Targeting human C-type lectin-like molecule-1 (CLL1) with a bispecific antibody for immunotherapy of acute myeloid leukemia. Angew. Chem. Int. Ed. Engl. 2014, 53, 9841–9845. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Uy, G.L.; Godwin, J.; Rettig, M.P.; Vey, N.; Foster, M.; Arellano, M.L.; Rizzieri, D.A.; Topp, M.S.; Huls, G.; Lowenberg, B. Preliminary results of a phase 1 study of flotetuzumab, a cd123 x cd3 bispecific dart® protein, in patients with relapsed/refractory acute myeloid leukemia and myelodysplastic syndrome. Blood 2017, 130, 637. [Google Scholar]
- Leong, S.R.; Sukumaran, S.; Hristopoulos, M.; Totpal, K.; Stainton, S.; Lu, E.; Wong, A.; Tam, L.; Newman, R.; Vuillemenot, B.R.; et al. An anti-CD3/anti-CLL-1 bispecific antibody for the treatment of acute myeloid leukemia. Blood 2017, 129, 609–618. [Google Scholar] [CrossRef] [Green Version]
- Busfield, S.J.; Biondo, M.; Wong, M.; Ramshaw, H.S.; Lee, E.M.; Ghosh, S.; Braley, H.; Panousis, C.; Roberts, A.W.; He, S.Z.; et al. Targeting of acute myeloid leukemia in vitro and in vivo with an anti-CD123 mAb engineered for optimal ADCC. Leukemia 2014, 28, 2213–2221. [Google Scholar] [CrossRef]
- Lee, E.M.; Yee, D.; Busfield, S.J.; McManus, J.F.; Cummings, N.; Vairo, G.; Wei, A.; Ramshaw, H.S.; Powell, J.A.; Lopez, A.F.; et al. Efficacy of an Fc-modified anti-CD123 antibody (CSL362) combined with chemotherapy in xenograft models of acute myelogenous leukemia in immunodeficient mice. Haematologica 2015, 100. [Google Scholar] [CrossRef]
- Smith, B.D.; Roboz, G.J.; Walter, R.B.; Altman, J.K.; Ferguson, A.; Curcio, T.J.; Orlowski, K.F.; Garrett, L.; Busfield, S.J.; Barnden, M.; et al. First-in Man, Phase 1 Study of CSL362 (Anti-IL3Rα / Anti-CD123 Monoclonal Antibody) in Patients with CD123+ Acute Myeloid Leukemia (AML) in CR at High Risk for Early Relapse. Blood 2014, 124, 120. [Google Scholar] [CrossRef]
- Kubasch, A.S.; Schulze, F.; Götze, K.S.; Krönke, J.; Sockel, K.; Middeke, J.M.; Chermat, F.; Gloaguen, S.; Puttrich, M.; Weigt, C.; et al. Anti-CD123 Targeted Therapy with Talacotuzumab in Advanced MDS and AML after Failing Hypomethylating Agents—Final Results of the Samba Trial. Blood 2018, 132, 4045. [Google Scholar] [CrossRef]
- Gleason, M.K.; Ross, J.A.; Warlick, E.D.; Lund, T.C.; Verneris, M.R.; Wiernik, A.; Spellman, S.; Haagenson, M.D.; Lenvik, A.J.; Litzow, M.R.; et al. CD16xCD33 bispecific killer cell engager (BiKE) activates NK cells against primary MDS and MDSC CD33+ targets. Blood 2014, 123, 3016–3026. [Google Scholar] [CrossRef]
- Vallera, D.A.; Felices, M.; McElmurry, R.; McCullar, V.; Zhou, X.; Schmohl, J.U.; Zhang, B.; Lenvik, A.J.; Panoskaltsis-Mortari, A.; Verneris, M.R.; et al. IL15 Trispecific Killer Engagers (TriKE) Make Natural Killer Cells Specific to CD33+ Targets While Also Inducing Persistence, In Vivo Expansion, and Enhanced Function. Clin. Cancer Res. 2016, 22, 3440–3450. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Kügler, M.; Stein, C.; Kellner, C.; Mentz, K.; Saul, D.; Schwenkert, M.; Schubert, I.; Singer, H.; Oduncu, F.; Stockmeyer, B.; et al. A recombinant trispecific single-chain Fv derivative directed against CD123 and CD33 mediates effective elimination of acute myeloid leukaemia cells by dual targeting. Br. J. Haematol. 2010, 150, 574–586. [Google Scholar] [CrossRef] [PubMed]
- Raneros, A.B.; Minguela, A.; Rodriguez, R.M.; Colado, E.; Bernal, T.; Anguita, E.; Mogorron, A.V.; Gil, A.C.; Vidal-Castiñeira, J.R.; Márquez-Kisinousky, L.; et al. Correction: Increasing TIMP3 expression by hypomethylating agents diminishes soluble MICA, MICB and ULBP2 shedding in acute myeloid leukemia, facilitating NK cell-mediated immune recognition. Oncotarget 2018, 9, 32881. [Google Scholar] [CrossRef] [PubMed]
- Herrmann, M.; Krupka, C.; Deiser, K.; Brauchle, B.; Marcinek, A.; Ogrinc, W.A.; Rataj, F.; Mocikat, R.; Metzeler, K.H.; Spiekermann, K.; et al. Bifunctional PD-1 × αCD3 × αCD33 fusion protein reverses adaptive immune escape in acute myeloid leukemia. Blood 2018, 132. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Schmohl, J.U.; Felices, M.; Todhunter, D.; Taras, E.; Miller, J.S.; Vallera, D.A. Tetraspecific scFv construct provides NK cell mediated ADCC and self-sustaining stimuli via insertion of IL-15 as a cross-linker. Oncotarget 2016, 7, 73830–73844. [Google Scholar] [CrossRef]
- Horowitz, M.M.; Gale, R.P.; Sondel, P.M.; Goldman, J.M.; Kersey, J.; Kolb, H.J.; Rimm, A.A.; Ringdén, O.; Rozman, C.; Speck, B. Graft-versus-leukemia reactions after bone marrow transplantation. Blood 1990, 75, 555–562. [Google Scholar] [CrossRef] [Green Version]
- Sakellari, I.; Konstantinou, V.; Gavriilaki, E.; Mallouri, D.; Batsis, I.; Kalaitzidou, V.; Apostolou, C.; Kaliou, M.; Bouinta, A.; Smias, C.; et al. Efficacy and Toxicity of Donor Lymphocyte Infusions in Relapsed Acute Myeloid Leukemia Post Allogeneic Hematopoietic Cell Transplantation: Single-Center Experience. Blood 2016, 128, 5804. [Google Scholar] [CrossRef]
- Mardiros, A.; Dos Santos, C.; McDonald, T.; Brown, C.E.; Wang, X.; Budde, L.E.; Hoffman, L.; Aguilar, B.; Chang, W.-C.; Bretzlaff, W.; et al. T cells expressing CD123-specific chimeric antigen receptors exhibit specific cytolytic effector functions and antitumor effects against human acute myeloid leukemia. Blood 2013, 122, 3138–3148. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Kenderian, S.S.; Ruella, M.; Shestova, O.; Klichinsky, M.; Aikawa, V.; Morrissette, J.J.D.; Scholler, J.; Song, D.; Porter, D.L.; 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]
- Petrov, J.C.; Wada, M.; Pinz, K.G.; Yan, L.E.; Chen, K.H.; Shuai, X.; Liu, H.; Chen, X.; Leung, L.-H.; Salman, H.; et al. Compound CAR T-cells as a double-pronged approach for treating acute myeloid leukemia. Leukemia 2018, 32, 1317–1326. [Google Scholar] [CrossRef] [Green Version]
- Brudno, J.N.; Kochenderfer, J.N. Toxicities of chimeric antigen receptor T cells: Recognition and management. Blood 2016, 127, 3321–3330. [Google Scholar] [CrossRef] [Green Version]
- Hattori, N.; Nakamaki, T. Natural Killer Immunotherapy for Minimal Residual Disease Eradication Following Allogeneic Hematopoietic Stem Cell Transplantation in Acute Myeloid Leukemia. Int. J. Mol. Sci. 2019, 20, 2057. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Xie, G.; Dong, H.; Liang, Y.; Ham, J.D.; Rizwan, R.; Chen, J. CAR-NK cells: A promising cellular immunotherapy for cancer. EBioMedicine 2020, 59, 102975. [Google Scholar] [CrossRef] [PubMed]
- 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]
- Tettamanti, S.; Marin, V.; Pizzitola, I.; Magnani, C.F.; Biagi, E. Targeting of acute myeloid leukemia (AML) by cytokine-induced killer (CIK) cells redirected with a novel CD123-specific chimeric antigen receptor (CAR). Br. J. Haematol. 2013, 161. [Google Scholar] [CrossRef]
- Bednarski, J.J.; Zimmerman, C.; Cashen, A.F.; Desai, S.; Foster, M.; Schappe, T.; McClain, E.; Becker-Hapak, M.; Berrien-Elliott, M.M.; Fehniger, T.A. Adoptively Transferred Donor-Derived Cytokine Induced Memory-like NK Cells Persist and Induce Remission in Pediatric Patient with Relapsed Acute Myeloid Leukemia after Hematopoietic Cell Transplantation. Blood 2019, 134, 3307. [Google Scholar] [CrossRef]
- 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]
- Curti, A.; Ruggeri, L.; D’Addio, A.; Bontadini, A.; Dan, E.; Motta, M.R.; Trabanelli, S.; Giudice, V.; Urbani, E.; Martinelli, G.; et al. Successful transfer of alloreactive haploidentical KIR ligand-mismatched natural killer cells after infusion in elderly high risk acute myeloid leukemia patients. Blood 2011, 118, 3273–3279. [Google Scholar] [CrossRef]
- 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. [Google Scholar] [CrossRef]
- Curti, A.; Ruggeri, L.; Parisi, S.; Bontadini, A.; Dan, E.; Motta, M.R.; Rizzi, S.; Trabanelli, S.; Ocadlikova, D.; Lecciso, M.; et al. 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, 1914–1921. [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]
- Ewen, E.-M.; Pahl, J.H.W.; Miller, M.; Watzl, C.; Cerwenka, A. KIR downregulation by IL-12/15/18 unleashes human NK cells from KIR/HLA-I inhibition and enhances killing of tumor cells. Eur. J. Immunol. 2018, 48, 355–365. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Ojo, E.O.; Sharma, A.A.; Liu, R.; Moreton, S.; Checkley-Luttge, M.-A.; Gupta, K.; Lee, G.; Lee, D.A.; Otegbeye, F.; Sekaly, R.-P.; et al. Membrane bound IL-21 based NK cell feeder cells drive robust expansion and metabolic activation of NK cells. Sci. Rep. 2019, 9, 14916. [Google Scholar] [CrossRef] [Green Version]
- 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]
- Leong, J.W.; Chase, J.M.; Romee, R.; Schneider, S.E.; Sullivan, R.P.; Cooper, M.A.; Fehniger, T.A. Preactivation with IL-12, IL-15, and IL-18 induces CD25 and a functional high-affinity IL-2 receptor on human cytokine-induced memory-like natural killer cells. Biol. Blood Marrow Transpl. 2014, 20, 463–473. [Google Scholar] [CrossRef] [Green Version]
- Buyse, M.; Squifflet, P.; Lange, B.J.; Alonzo, T.A.; Larson, R.A.; Kolitz, J.E.; George, S.L.; Bloomfield, C.D.; Castaigne, S.; Chevret, S.; et al. Individual patient data meta-analysis of randomized trials evaluating IL-2 monotherapy as remission maintenance therapy in acute myeloid leukemia. Blood 2011, 117, 7007–7013. [Google Scholar] [CrossRef] [PubMed]
- Lange, B.J.; Yang, R.K.; Gan, J.; Hank, J.A.; Sievers, E.L.; Alonzo, T.A.; Gerbing, R.B.; Sondel, P.M. Soluble interleukin-2 receptor α activation in a Children’s Oncology Group randomized trial of interleukin-2 therapy for pediatric acute myeloid leukemia. Pediatr. Blood Cancer 2011, 57, 398–405. [Google Scholar] [CrossRef] [Green Version]
- Mao, C.; Fu, X.-H.; Yuan, J.-Q.; Yang, Z.-Y.; Huang, Y.-F.; Ye, Q.-L.; Wu, X.-Y.; Hu, X.-F.; Zhai, Z.-M.; Tang, J.-L. Interleukin-2 as maintenance therapy for children and adults with acute myeloid leukaemia in first complete remission. Cochrane Database Syst. Rev. 2015, CD010248. [Google Scholar] [CrossRef]
- 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]
- Soiffer, R.J. Can IL-15 superagonist ALTer GVL? Blood 2018, 131, 2511–2512. [Google Scholar] [CrossRef]
- Vasu, S.; Sharma, N.; Odonnell, L.; Bosse, K.; Lee, D.A. A phase I clinical trial testing the safety of IL-21-expanded, off-the-shelf, natural killer cells for relapsed/refractory acute myeloid leukemia and myelodysplastic syndrome. J. Clin. Oncol. 2020, 38, TPS7562. [Google Scholar] [CrossRef]
- 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]
- Anguille, S.; Lion, E.; Willemen, Y.; van Tendeloo, V.F.I.; Berneman, Z.N.; Smits, E.L.J.M. Interferon-α in acute myeloid leukemia: An old drug revisited. Leukemia 2011, 25, 739–748. [Google Scholar] [CrossRef] [Green Version]
- Smits, E.L.J.M.; Anguille, S.; Berneman, Z.N. Interferon α may be back on track to treat acute myeloid leukemia. Oncoimmunology 2013, 2, e23619. [Google Scholar] [CrossRef] [Green Version]
- John, F. Foley. New connections: TGF-β in tumors. Sci. Signal. 2018, 11. [Google Scholar] [CrossRef]
- Otegbeye, F.; Ojo, E.; Moreton, S.; Mackowski, N.; Lee, D.A.; De Lima, M.; Wald, D.N. Inhibiting TGF-beta signaling preserves the function of highly activated, in vitro expanded natural killer cells in AML and colon cancer models. PLoS ONE 2018, 13, e0191358. [Google Scholar] [CrossRef] [Green Version]
- Oka, Y.; Tsuboi, A.; Taguchi, T.; Osaki, T.; Kyo, T.; Nakajima, H.; Elisseeva, O.A.; Oji, Y.; Kawakami, M.; Ikegame, K.; et al. Induction of WT1 (Wilms’ tumor gene)-specific cytotoxic T lymphocytes by WT1 peptide vaccine and the resultant cancer regression. Proc. Natl. Acad. Sci. USA 2004, 101. [Google Scholar] [CrossRef] [Green Version]
- Maeda, T.; Hosen, N.; Fukushima, K.; Tsuboi, A.; Morimoto, S.; Matsui, T.; Sata, H.; Fujita, J.; Hasegawa, K.; Nishida, S.; et al. Maintenance of complete remission after allogeneic stem cell transplantation in leukemia patients treated with Wilms tumor 1 peptide vaccine. Blood Cancer J. 2013, 3, e130. [Google Scholar] [CrossRef] [PubMed]
- Maslak, P.G.; Dao, T.; Bernal, Y.; Chanel, S.M.; Zhang, R.; Frattini, M.; Rosenblat, T.; Jurcic, J.G.; Brentjens, R.J.; Arcila, M.E.; et al. Phase 2 trial of a multivalent WT1 peptide vaccine (galinpepimut-S) in acute myeloid leukemia. Blood Adv. 2018, 2. [Google Scholar] [CrossRef]
- Anguille, S.; van de Velde, A.L.; Smits, E.L.; van Tendeloo, V.F.; Juliusson, G.; Cools, N.; Nijs, G.; Stein, B.; Lion, E.; van Driessche, A.; et al. Dendritic cell vaccination as postremission treatment to prevent or delay relapse in acute myeloid leukemia. Blood 2017, 130, 1713–1721. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Khoury, H.J.; Collins, R.H.; Blum, W.; Stiff, P.S.; Elias, L.; Lebkowski, J.S.; Reddy, A.; Nishimoto, K.P.; Sen, D.; Wirth, E.D.; et al. Immune responses and long-term disease recurrence status after telomerase-based dendritic cell immunotherapy in patients with acute myeloid leukemia. Cancer 2017, 123, 3061–3072. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Rosenblatt, J.; Stone, R.M.; Uhl, L.; Neuberg, D.; Joyce, R.; Levine, J.D.; Arnason, J.; McMasters, M.; Luptakova, K.; Jain, S.; et al. Individualized vaccination of AML patients in remission is associated with induction of antileukemia immunity and prolonged remissions. Sci. Transl. Med. 2016, 8, 368ra171. [Google Scholar] [CrossRef] [Green Version]
- Shah, N.J.; Najibi, A.J.; Shih, T.-Y.; Mao, A.S.; Sharda, A.; Scadden, D.T.; Mooney, D.J. A biomaterial-based vaccine eliciting durable tumour-specific responses against acute myeloid leukaemia. Nat. Biomed. Eng. 2020, 4, 40–51. [Google Scholar] [CrossRef]
- Russell, L.; Peng, K.W.; Russell, S.J.; Diaz, R.M. Oncolytic Viruses: Priming Time for Cancer Immunotherapy. BioDrugs 2019, 33, 485–501. [Google Scholar] [CrossRef] [Green Version]
- Adair, R.A.; Roulstone, V.; Scott, K.J.; Morgan, R.; Nuovo, G.J.; Fuller, M.; Beirne, D.; West, E.J.; Jennings, V.A.; Rose, A.; et al. Cell carriage, delivery, and selective replication of an oncolytic virus in tumor in patients. Sci. Transl. Med. 2012, 4, 138ra77. [Google Scholar] [CrossRef] [Green Version]
- Kaufman, H.L.; Kohlhapp, F.J.; Zloza, A. Oncolytic viruses: A new class of immunotherapy drugs. Nat. Rev. Drug Discov. 2015, 14, 642–662. [Google Scholar] [CrossRef]
- Shi, T.; Song, X.; Wang, Y.; Liu, F.; Wei, J. Combining Oncolytic Viruses with Cancer Immunotherapy: Establishing a New Generation of Cancer Treatment. Front. Immunol. 2020, 11, 683. [Google Scholar] [CrossRef]
- Maurer, S.; Salih, H.R.; Smirnow, I.; Lauer, U.M.; Berchtold, S. Suicide gene-armed measles vaccine virus for the treatment of AML. Int. J. Oncol. 2019, 55, 347–358. [Google Scholar] [CrossRef]
- Wang, Z.; Liu, W.; Wang, L.; Gao, P.; Li, Z.; Wu, J.; Zhang, H.; Wu, H.; Kong, W.; Yu, B.; et al. Enhancing the antitumor activity of an engineered TRAIL-coated oncolytic adenovirus for treating acute myeloid leukemia. Signal. Transduct. Target. Ther. 2020, 5, 40. [Google Scholar] [CrossRef] [PubMed]
- Samudio, I.; Rezvani, K.; Shaim, H.; Hofs, E.; Ngom, M.; Bu, L.; Liu, G.; Lee, J.T.C.; Imren, S.; Lam, V.; et al. UV-inactivated HSV-1 potently activates NK cell killing of leukemic cells. Blood 2016, 127, 2575–2586. [Google Scholar] [CrossRef] [Green Version]
- Müller, L.M.E.; Holmes, M.; Michael, J.L.; Scott, G.B.; West, E.J.; Scott, K.J.; Parrish, C.; Hall, K.; Stäble, S.; Jennings, V.A.; et al. Plasmacytoid dendritic cells orchestrate innate and adaptive anti-tumor immunity induced by oncolytic coxsackievirus A21. J. Immunother. Cancer 2019, 7, 164. [Google Scholar] [CrossRef] [Green Version]
- Hall, K.; Scott, K.J.; Rose, A.; Desborough, M.; Harrington, K.; Pandha, H.; Parrish, C.; Vile, R.; Coffey, M.; Bowen, D.; et al. Reovirus-mediated cytotoxicity and enhancement of innate immune responses against acute myeloid leukemia. Biores. Open Access 2012, 1, 3–15. [Google Scholar] [CrossRef]
- Pol, J.G.; Zhang, L.; Bridle, B.W.; Stephenson, K.B.; Rességuier, J.; Hanson, S.; Chen, L.; Kazdhan, N.; Bramson, J.L.; Stojdl, D.F.; et al. Maraba virus as a potent oncolytic vaccine vector. Mol. Ther. 2014, 22, 420–429. [Google Scholar] [CrossRef] [Green Version]
- Bridle, B.W.; Boudreau, J.E.; Lichty, B.D.; Brunellière, J.; Stephenson, K.; Koshy, S.; Bramson, J.L.; Wan, Y. Vesicular Stomatitis Virus as a Novel Cancer Vaccine Vector to Prime Antitumor Immunity Amenable to Rapid Boosting with Adenovirus. Mol. Ther. 2009, 17, 1814–1821. [Google Scholar] [CrossRef] [PubMed]
- Shen, W.; Patnaik, M.M.; Ruiz, A.; Russell, S.J.; Peng, K.-W. Immunovirotherapy with vesicular stomatitis virus and PD-L1 blockade enhances therapeutic outcome in murine acute myeloid leukemia. Blood 2016, 127, 1449–1458. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Pol, J.G.; Workenhe, S.T.; Konda, P.; Gujar, S.; Kroemer, G. Cytokines in oncolytic virotherapy. Cytokine Growth Factor Rev. 2020, 56, 4–27. [Google Scholar] [CrossRef]
- Guo, Z.S.; Lotze, M.T.; Zhu, Z.; Storkus, W.J.; Song, X.-T. Bi- and Tri-Specific T Cell Engager-Armed Oncolytic Viruses: Next-Generation Cancer Immunotherapy. Biomedicines 2020, 8, 204. [Google Scholar] [CrossRef]
- Park, A.K.; Fong, Y.; Kim, S.-I.; Yang, J.; Murad, J.P.; Lu, J.; Jeang, B.; Chang, W.-C.; Chen, N.G.; Thomas, S.H.; et al. Effective combination immunotherapy using oncolytic viruses to deliver CAR targets to solid tumors. Sci. Transl. Med. 2020, 12. [Google Scholar] [CrossRef] [PubMed]
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
Sendker, S.; Reinhardt, D.; Niktoreh, N. Redirecting the Immune Microenvironment in Acute Myeloid Leukemia. Cancers 2021, 13, 1423. https://doi.org/10.3390/cancers13061423
Sendker S, Reinhardt D, Niktoreh N. Redirecting the Immune Microenvironment in Acute Myeloid Leukemia. Cancers. 2021; 13(6):1423. https://doi.org/10.3390/cancers13061423
Chicago/Turabian StyleSendker, Stephanie, Dirk Reinhardt, and Naghmeh Niktoreh. 2021. "Redirecting the Immune Microenvironment in Acute Myeloid Leukemia" Cancers 13, no. 6: 1423. https://doi.org/10.3390/cancers13061423
APA StyleSendker, S., Reinhardt, D., & Niktoreh, N. (2021). Redirecting the Immune Microenvironment in Acute Myeloid Leukemia. Cancers, 13(6), 1423. https://doi.org/10.3390/cancers13061423