Emerging Immunotherapy for Acute Myeloid Leukemia
Abstract
:1. Introduction
2. Molecules Involved in IO Therapy
2.1. Immune Checkpoint Molecules
2.1.1. Programmed Cell Death 1 (PD-1), Programmed Cell Death-Ligand 1 (PD-L1), and Cytotoxic T-Lymphocyte-Associated Protein 4 (CTLA-4)
2.1.2. T-Cell Immunoglobulin and Mucin-Domain Containing-3 (TIM-3)
2.1.3. Lymphocyte Activation Gene-3 Protein (LAG-3)
2.1.4. Leukocyte Surface Antigen CD47
2.1.5. Other Checkpoint Molecules
2.2. Potential Immune Targets on Leukemic Cells
2.2.1. Interleukin-3 Receptor Subunit Alpha (IL-3RA) or CD123
2.2.2. Myeloid Cell Surface Antigen CD33
2.2.3. C-Type Lectin-Like Molecule-1 (CLL-1)
2.2.4. Other Candidates of Immune Targets
3. Novel IO Therapy in Clinical Trials
3.1. Immune Checkpoint Inhibitors
3.1.1. Anti-PD-1/CTLA-4 Antibody
3.1.2. Anti-TIM-3 Antibody
3.2. CAR-T Therapy and Its Relatives
3.3. Bispecific and Trispecific Antibodies: BiTE and DART
3.4. Antibody-Drug Conjugate
4. Conclusions
Author Contributions
Funding
Institutional Review Board Statement
Informed Consent Statement
Data Availability Statement
Conflicts of Interest
References
- Specenier, P. Ipilimumab in melanoma. Expert Rev. Anticancer Ther. 2016, 16, 811–826. [Google Scholar] [CrossRef]
- Kantarjian, H.M.; Stein, A.; Gökbuget, N.; Fielding, A.K.; Schuh, A.C.; Ribera, J.-M.; Wei, A.; Dombret, H.; Foà, R.; Bassan, R.; et al. Blinatumomab versus Chemotherapy for Advanced Acute Lymphoblastic Leukemia. N. Engl. J. Med. 2017, 376, 836–847. [Google Scholar] [CrossRef] [PubMed]
- Goebeler, M.-E.; Knop, S.; Viardot, A.; Kufer, P.; Topp, M.S.; Einsele, H.; Noppeney, R.; Hess, G.; Kallert, S.; Mackensen, A.; et al. Bispecific T-Cell Engager (BiTE) Antibody Construct Blinatumomab for the Treatment of Patients With Relapsed/Refractory Non-Hodgkin Lymphoma: Final Results From a Phase I Study. J. Clin. Oncol. 2016, 34, 1104–1111. [Google Scholar] [CrossRef]
- Viardot, A.; Goebeler, M.-E.; Hess, G.; Neumann, S.; Pfreundschuh, M.; Adrian, N.; Zettl, F.; Libicher, M.; Sayehli, C.; Stieglmaier, J.; et al. Phase 2 study of the bispecific T-cell engager (BiTE) antibody blinatumomab in relapsed/refractory diffuse large B-cell lymphoma. Blood 2016, 127, 1410–1416. [Google Scholar] [CrossRef] [PubMed]
- 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]
- Park, J.H.; Rivière, I.; Gonen, M.; Wang, X.; Sénéchal, B.; Curran, K.J.; Sauter, C.; Wang, Y.; Santomasso, B.; Mead, E.; et al. Long-Term Follow-up of CD19 CAR Therapy in Acute Lymphoblastic Leukemia. N. Engl. J. Med. 2018, 378, 449–459. [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] [PubMed]
- Chevallier, P.; Delaunay, J.; Turlure, P.; Pigneux, A.; Hunault, M.; Garand, R.; Guillaume, T.; Avet-Loiseau, H.; Dmytruk, N.; Girault, S.; et al. Long-Term Disease-Free Survival After Gemtuzumab, Intermediate-Dose Cytarabine, and Mitoxantrone in Patients With CD33+Primary Resistant or Relapsed Acute Myeloid Leukemia. J. Clin. Oncol. 2008, 26, 5192–5197. [Google Scholar] [CrossRef]
- 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]
- Fife, B.T.; Pauken, K.E. The role of the PD-1 pathway in autoimmunity and peripheral tolerance. Ann. N. Y. Acad. Sci. 2011, 1217, 45–59. [Google Scholar] [CrossRef]
- Berger, K.N.; Pu, J.J. PD-1 pathway and its clinical application: A 20 year journey after discovery of the complete human PD-1 gene. Gene 2018, 638, 20–25. [Google Scholar] [CrossRef]
- Linsley, P.S.; Brady, W.; Urnes, M.; Grosmaire, L.S.; Damle, N.K.; A Ledbetter, J. CTLA-4 is a second receptor for the B cell activation antigen B7. J. Exp. Med. 1991, 174, 561–569. [Google Scholar] [CrossRef] [Green Version]
- Teft, W.A.; Kirchhof, M.G.; Madrenas, J. A MOLECULAR PERSPECTIVE OF CTLA-4 FUNCTION. Annu. Rev. Immunol. 2006, 24, 65–97. [Google Scholar] [CrossRef] [Green Version]
- LaBelle, J.L.; Hanke, C.A.; Blazar, B.R.; Truitt, R.L. Negative effect of CTLA-4 on induction of T-cell immunity in vivo to B7-1+, but not B7-2+, murine myelogenous leukemia. Blood J. Am. Soc. Hematol. 2002, 99, 2146–2153. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Yang, H.; Bueso-Ramos, C.; Dinardo, C.D.; 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] [PubMed]
- Chen, X.; Liu, S.; Wang, L.; Zhang, W.-G.; Ji, Y.; Ma, X. Clinical significance of B7-H1 (PD-L1) expression in human acute leukemia. Cancer Biol. Ther. 2008, 7, 622–627. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Goltz, D.; Gevensleben, H.; Grünen, S.; Dietrich, J.; Kristiansen, G.; Landsberg, J. PD-L1 (CD274) promoter methylation predicts survival in patients with acute myeloid leukemia. Leukemia 2017, 31, 738–743. [Google Scholar] [CrossRef] [PubMed]
- Goswami, M.; Oetjen, K.; Mulé, B.M.P.; Sheela, M.S.; Wong, H.Y.; Liu, Q.; Calvo, K.R.; Lai, C.E.; Hourigan, C.S. Increased Frequencies of PD-1+ CD8+ Marrow-Infiltrating Lymphocytes Associated with Highly Clonal T-Lymphocyte Expansions in Relapsed and Refractory AML Patients but Not Healthy Adults. Blood 2016, 128, 1644. [Google Scholar] [CrossRef]
- Daver, N.; Basu, S.; Garcia-Manero, G.; Cortes, J.E.; Ravandi, F.; Ning, J.; Xiao, L.; Juliana, L.; Kornblau, S.M.; Konopleva, M.; et al. Defining the Immune Checkpoint Landscape in Patients (pts) with Acute Myeloid Leukemia (AML). Blood 2016, 128, 2900. [Google Scholar] [CrossRef]
- Tan, J.; Chen, S.; Lu, Y.; Yao, D.; Xu, L.; Zhang, Y.; Yang, L.; Chen, J.; Lai, J.; Yu, Z.; et al. Higher PD-1 expression concurrent with exhausted CD8+ T cells in patients with de novo acute myeloid leukemia. Chin. J. Cancer Res. 2017, 29, 463–470. [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, 93. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Monney, L.; Sabatos, C.A.; Gaglia, J.L.; Ryu, A.; Waldner, H.; Chernova, T.; Manning, S.; Greenfield, E.A.; Coyle, A.J.; Sobel, R.A.; et al. Th1-specific cell surface protein Tim-3 regulates macrophage activation and severity of an autoimmune disease. Nat. Cell Biol. 2002, 415, 536–541. [Google Scholar] [CrossRef]
- Sánchez-Fueyo, A.; Tian, J.; Picarella, D.; Domenig, C.; Zheng, X.X.; Sabatos, C.A.; Manlongat, N.; Bender, O.; Kamradt, T.; Kuchroo, V.K.; et al. Tim-3 inhibits T helper type 1-mediated auto- and alloimmune responses and promotes immunological tolerance. Nat Immunol. 2003, 4, 1093–1101. [Google Scholar] [CrossRef] [PubMed]
- Tomkowicz, B.; Walsh, E.R.; Cotty, A.; Verona, R.; Sabins, N.; Kaplan, F.; Santulli-Marotto, S.; Chin, C.-N.; Mooney, J.L.; Lingham, R.B.; et al. TIM-3 Suppresses Anti-CD3/CD28-Induced TCR Activation and IL-2 Expression through the NFAT Signaling Pathway. PLoS ONE 2015, 10, e0140694. [Google Scholar] [CrossRef] [Green Version]
- Gautron, A.-S.; Dominguez-Villar, M.; De Marcken, M.; Hafler, D.A. Enhanced suppressor function of TIM-3+FoxP3+regulatory T cells. Eur. J. Immunol. 2014, 44, 2703–2711. [Google Scholar] [CrossRef] [Green Version]
- Wang, F.; He, W.; Zhou, H.; Yuan, J.; Wu, K.; Xu, L.; Chen, Z.K. The Tim-3 ligand galectin-9 negatively regulates CD8+ alloreactive T cell and prolongs survival of skin graft. Cell. Immunol. 2007, 250, 68–74. [Google Scholar] [CrossRef]
- Bi, S.; Hong, P.W.; Lee, B.; Baum, L.G. Galectin-9 binding to cell surface protein disulfide isomerase regulates the redox environment to enhance T-cell migration and HIV entry. Proc. Natl. Acad. Sci. USA 2011, 108, 10650–10655. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Gieseke, F.; Kruchen, A.; Tzaribachev, N.; Bentzien, F.; Dominici, M.; Müller, I. Proinflammatory stimuli induce galectin-9 in human mesenchymal stromal cells to suppress T-cell proliferation. Eur. J. Immunol. 2013, 43, 2741–2749. [Google Scholar] [CrossRef] [Green Version]
- Golden-Mason, L.; Mcmahan, R.H.; Strong, M.; Reisdorph, R.; Mahaffey, S.; Palmer, B.E.; Cheng, L.; Kulesza, C.; Hirashima, M.; Niki, T.; et al. Galectin-9 Functionally Impairs Natural Killer Cells in Humans and Mice. J. Virol. 2013, 87, 4835–4845. [Google Scholar] [CrossRef] [Green Version]
- DeKruyff, R.H.; Bu, X.; Ballesteros, A.; Santiago, C.; Chim, Y.-L.E.; Lee, H.-H.; Karisola, P.; Pichavant, M.; Kaplan, G.G.; Umetsu, D.T.; et al. T Cell/Transmembrane, Ig, and Mucin-3 Allelic Variants Differentially Recognize Phosphatidylserine and Mediate Phagocytosis of Apoptotic Cells. J. Immunol. 2010, 184, 1918–1930. [Google Scholar] [CrossRef] [Green Version]
- Chiba, S.; Baghdadi, M.; Akiba, H.; Yoshiyama, H.; Kinoshita, I.; Dosaka-Akita, H.; Fujioka, Y.; Ohba, Y.; Gorman, J.V.; Colgan, J.D.; et al. Tumor-infiltrating DCs suppress nucleic acid–mediated innate immune responses through interactions between the receptor TIM-3 and the alarmin HMGB1. Nat. Immunol. 2012, 13, 832–842. [Google Scholar] [CrossRef]
- Huang, Y.-H.; Zhu, C.; Kondo, Y.; Anderson, A.C.; Gandhi, A.; Russell, A.F.; Dougan, S.K.; Petersen, B.-S.; Melum, E.; Pertel, T.; et al. CEACAM1 regulates TIM-3-mediated tolerance and exhaustion. Nat. Cell Biol. 2015, 517, 386–390. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Li, C.; Chen, X.; Yu, X.; Zhu, Y.; Ma, C.; Xia, R.; Ma, J.; Gu, C.; Ye, L.; Wu, D. Tim-3 is highly expressed in T cells in acute myeloid leukemia and associated with clinicopathological prognostic stratification. Int. J. Clin. Exp. Pathol. 2014, 7, 6880–6888. [Google Scholar]
- Kong, Y.; Zhang, J.; Claxton, D.F.; Ehmann, W.C.; Rybka, W.B.; Zhu, L.; Zeng, H.; Schell, T.D.; Zheng, H. PD-1(hi)TIM-3(+) T cells associate with and predict leukemia relapse in AML patients post allogeneic stem cell transplantation. Blood Cancer J. 2015, 5, e330. [Google Scholar] [CrossRef] [Green Version]
- 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 J. Am. Soc. Hematol. 2011, 117, 10. [Google Scholar]
- Han, G.; Chen, G.; Shen, B.; Li, Y. Tim-3: An Activation Marker and Activation Limiter of Innate Immune Cells. Front. Immunol. 2013, 4. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Gleason, M.K.; Lenvik, T.R.; McCullar, V.; Felices, M.; O’Brien, M.S.; Cooley, S.A.; Verneris, M.R.; Cichocki, F.; Holman, C.J.; Panoskaltsis-Mortari, A.; et al. Tim-3 is an inducible human natural killer cell receptor that enhances interferon gamma production in response to galectin-9. Blood 2012, 119, 3064–3072. [Google Scholar] [CrossRef]
- Folgiero, V.; Cifaldi, L.; Pira, G.L.; Goffredo, B.M.; Vinti, L.; Locatelli, F. TIM-3/Gal-9 interaction induces IFNγ-dependent IDO1 expression in acute myeloid leukemia blast cells. J. Hematol. Oncol. 2015, 8, 1–5. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Silva, I.G.; 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, 44–57. [Google Scholar] [CrossRef] [Green Version]
- He, X.; Feng, Z.; Ma, J.; Ling, S.; Cao, Y.; Gurung, B.; Wu, Y.; Katona, B.W.; O’Dwyer, K.P.; Siegel, D.L.; et al. Bispecific and split CAR T cells targeting CD13 and TIM3 eradicate acute myeloid leukemia. Blood 2020, 135, 713–723. [Google Scholar] [CrossRef]
- Baixeras, E.; Huard, B.; Miossec, C.; Jitsukawa, S.; Martin, M.; Hercend, T.; Auffray, C.; Triebel, F.; Piatier-Tonneau, D. Characterization of the lymphocyte activation gene 3-encoded protein. A new ligand for human leukocyte antigen class II antigens. J. Exp. Med. 1992, 176, 327–337. [Google Scholar] [CrossRef] [PubMed]
- Workman, C.J.; Wang, Y.; El Kasmi, K.C.; Pardoll, D.M.; Murray, P.J.; Drake, C.G.; Vignali, D.A.A. LAG-3 Regulates Plasmacytoid Dendritic Cell Homeostasis. J. Immunol. 2009, 182, 1885–1891. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Camisaschi, C.; Casati, C.; Rini, F.; Perego, M.; De Filippo, A.; Triebel, F.; Parmiani, G.; Belli, F.; Rivoltini, L.; Castelli, C. LAG-3 Expression Defines a Subset of CD4+CD25highFoxp3+ Regulatory T Cells That Are Expanded at Tumor Sites. J. Immunol. 2010, 184, 6545–6551. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Triebel, F.; Jitsukawa, S.; Baixeras, E.; Roman-Roman, S.; Genevee, C.; Viegas-Pequignot, E.; Hercend, T. LAG-3, a novel lymphocyte activation gene closely related to CD4. J. Exp. Med. 1990, 171, 1393–1405. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Huard, B.; Tournier, M.; Hercend, T.; Triebel, F.; Faure, F. Lymphocyte-activation gene 3/major histocompatibility complex class II interaction modulates the antigenic response of CD4+ T lymphocytes. Eur. J. Immunol. 1994, 24, 3216–3221. [Google Scholar] [CrossRef] [PubMed]
- Huard, B.; Prigent, P.; Pagès, F.; Bruniquel, D.; Triebel, F. T cell major histocompatibility complex class II molecules down-regulate CD4+ T cell clone responses following LAG-3 binding. Eur. J. Immunol. 1996, 26, 1180–1186. [Google Scholar] [CrossRef]
- Woo, S.-R.; Turnis, M.E.; Goldberg, M.V.; Bankoti, J.; Selby, M.; Nirschl, C.J.; Bettini, M.L.; Gravano, D.M.; Vogel, P.; Liu, C.L.; et al. Immune Inhibitory Molecules LAG-3 and PD-1 Synergistically Regulate T-cell Function to Promote Tumoral Immune Escape. Cancer Res. 2012, 72, 917–927. [Google Scholar] [CrossRef] [Green Version]
- Okazaki, T.; Okazaki, I.-M.; Wang, J.; Sugiura, D.; Nakaki, F.; Yoshida, T.; Kato, Y.; Fagarasan, S.; Muramatsu, M.; Eto, T.; et al. PD-1 and LAG-3 inhibitory co-receptors act synergistically to prevent autoimmunity in mice. J. Exp. Med. 2011, 208, 395–407. [Google Scholar] [CrossRef] [Green Version]
- 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]
- Wierz, M.; Pierson, S.; Guyonnet, L.; Viry, E.; Lequeux, A.; Oudin, A.; Niclou, S.P.; Ollert, M.; Berchem, G.; Janji, B.; et al. Dual PD1/LAG3 immune checkpoint blockade limits tumor development in a murine model of chronic lymphocytic leukemia. Blood 2018, 131, 1617–1621. [Google Scholar] [CrossRef] [Green Version]
- Bristol-Myers Squibb. A Phase 1/2a Dose Escalation and Cohort Expansion Study of the Safety, Tolerability, and Efficacy of Anti-LAG-3 (BMS-986016) in Monoclonal Antibody (BMS-986016) Administered Alone and in Combination With Anti-PD-1 Monoclonal Antibody (Nivolumab, BMS-936558) in Relapsed or Refractory B-Cell Malignancies. clinicaltrials.gov. 2019. Available online: https://clinicaltrials.gov/ct2/show/NCT02061761 (accessed on 21 June 2020).
- Bristol-Myers Squibb. A Phase I/2a Dose Escalation and Cohort Expansion Study of the Safety, Tolerability, and Efficacy of Anti-LAG-3 Monoclonal Antibody (BMS-986016) Administered Alone and in Combination With Anti-PD-1 Monoclonal Antibody (Nivolumab, BMS-936558) in Advanced Solid Tumors. clinicaltrials.gov. 2020. Available online: https://clinicaltrials.gov/ct2/show/NCT01968109 (accessed on 21 June 2020).
- Latour, S.; Tanaka, H.; Demeure, C.; Mateo, V.; Rubio, M.; Brown, E.J.; Maliszewski, C.; Lindberg, F.P.; Oldenborg, A.; Ullrich, A.; et al. Bidirectional Negative Regulation of Human T and Dendritic Cells by CD47 and Its Cognate Receptor Signal-Regulator Protein-α: Down-Regulation of IL-12 Responsiveness and Inhibition of Dendritic Cell Activation. J. Immunol. 2001, 167, 2547–2554. [Google Scholar] [CrossRef] [Green Version]
- Oldenborg, P.-A.; Gresham, H.D.; Lindberg, F.P. Cd47-Signal Regulatory Protein α (Sirpα) Regulates Fcγ and Complement Receptor–Mediated Phagocytosis. J. Exp. Med. 2001, 193, 855–862. [Google Scholar] [CrossRef] [PubMed]
- Piccio, L.; Vermi, W.; Boles, K.S.; Fuchs, A.; Strader, C.A.; Facchetti, F.; Cella, M.; Colonna, M. Adhesion of human T cells to antigen-presenting cells through SIRP2-CD47 interaction costimulates T-cell proliferation. Blood 2005, 105, 7. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Jaiswal, S.; Jamieson, C.H.; Pang, W.W.; Park, C.Y.; Chao, M.P.; Majeti, R.; Traver, D.; Van Rooijen, N.; Weissman, I.L. CD47 Is Upregulated on Circulating Hematopoietic Stem Cells and Leukemia Cells to Avoid Phagocytosis. Cell 2009, 138, 271–285. [Google Scholar] [CrossRef] [Green Version]
- Majeti, R.; Chao, M.P.; Alizadeh, A.A.; Pang, W.W.; Jaiswal, S.; Gibbs, K.D.; Van Rooijen, N.; Weissman, I.L. CD47 Is an Adverse Prognostic Factor and Therapeutic Antibody Target on Human Acute Myeloid Leukemia Stem Cells. Cell 2009, 138, 286–299. [Google Scholar] [CrossRef] [Green Version]
- Petrova, P.S.; Viller, N.N.; Wong, M.; Pang, X.; Lin, G.H.Y.; Dodge, K.; Chai, V.; Chen, H.; Lee, V.; House, V.; et al. TTI-621 (SIRPαFc): A CD47-Blocking Innate Immune Checkpoint Inhibitor with Broad Antitumor Activity and Minimal Erythrocyte Binding. Clin. Cancer Res. 2017, 23, 1068–1079. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Advani, R.; Flinn, I.; Popplewell, L.; Forero, A.; Bartlett, N.L.; Ghosh, N.; Kline, J.; Roschewski, M.; LaCasce, A.; Collins, G.P.; et al. CD47 Blockade by Hu5F9-G4 and Rituximab in Non-Hodgkin’s Lymphoma. N. Engl. J. Med. 2018, 379, 1711–1721. [Google Scholar] [CrossRef]
- Brierley, C.; Staves, J.; Roberts, C.; Johnson, H.; Vyas, P.; Goodnough, L.; Murphy, M. The effects of monoclonal anti-CD47 on RBCs, compatibility testing, and transfusion requirements in refractory acute myeloid leukemia. Transfusion 2019, 59, 2248–2254. [Google Scholar] [CrossRef]
- Hobo, W.; Hutten, T.J.A.; Schaap, N.P.M.; Dolstra, H. Immune checkpoint molecules in acute myeloid leukaemia: Managing the double-edged sword. Br. J. Haematol. 2018, 181, 38–53. [Google Scholar] [CrossRef] [Green Version]
- Krieg, C.; Han, P.; Stone, R.; Goularte, O.D.; Kaye, J. Functional Analysis of B and T Lymphocyte Attenuator Engagement on CD4+ and CD8+ T Cells. J. Immunol. 2005, 175, 6420–6427. [Google Scholar] [CrossRef] [Green Version]
- Wright, G.J.; Puklavec, M.J.; Willis, A.C.; Hoek, R.M.; Sedgwick, J.D.; Brown, M.H.; Barclay, A. Lymphoid/Neuronal Cell Surface OX2 Glycoprotein Recognizes a Novel Receptor on Macrophages Implicated in the Control of Their Function. Immunity 2000, 13, 233–242. [Google Scholar] [CrossRef] [Green Version]
- Chen, J.-X.; Mei, L.-P.; Chen, B.-G.; Wang, D.-L.; Luo, W.-D.; Luo, L.-F.; Lu, R.; Zheng, R.; Zhang, L. Over-expression of CD200 predicts poor prognosis in MDS. Leuk. Res. 2017, 56, 1–6. [Google Scholar] [CrossRef] [PubMed]
- Lasaro, M.; Sazanovich, M.; Giles-Davis, W.; Mrass, P.; Bunte, R.M.; Sewell, D.; Hussain, S.F.; Fu, Y.-X.; Weninger, W.; Paterson, Y.; et al. Active Immunotherapy Combined With Blockade of a Coinhibitory Pathway Achieves Regression of Large Tumor Masses in Cancer-prone Mice. Mol. Ther. 2011, 19, 1727–1736. [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]
- Prasad, D.V.R.; Nguyen, T.; Li, Z.; Yang, Y.; Duong, J.; Wang, Y.; Dong, C. Murine B7-H3 is a negative regulator of T cells. J. Immunol. 2004, 173, 2500–2506. [Google Scholar] [CrossRef] [Green Version]
- Picarda, E.; Ohaegbulam, K.C.; Zang, X. Molecular Pathways: Targeting B7-H3 (CD276) for Human Cancer Immunotherapy. Clin. Cancer Res. 2016, 22, 3425–3431. [Google Scholar] [CrossRef] [Green Version]
- Lichtman, E.; Du, H.; Savoldo, B.; Ferrone, S.; Li, G.; Su, L.; Dotti, G. Pre-Clinical Evaluation of B7-H3-Specific Chimeric Antigen Receptor T-Cells for the Treatment of Acute Myeloid Leukemia. Blood 2018, 132, 701. [Google Scholar] [CrossRef]
- Muñoz, L.; Nomdedéu, J.F.; López, O.; Carnicer, M.J.; Bellido, M.; Aventín, A.; Brunet, S.; Sierra, J. Interleukin-3 receptor alpha chain (CD123) is widely expressed in hematologic malignancies. Haematologica 2001, 86, 1261–1269. [Google Scholar]
- Testa, U.; Pelosi, E.; Frankel, A. CD 123 is a membrane biomarker and a therapeutic target in hematologic malignancies. Biomark. Res. 2014, 2, 4. [Google Scholar] [CrossRef] [Green Version]
- Ehninger, G.; on behalf of the Study Alliance Leukemia; Kramer, M.; Rollig, C.; Thiede, C.; Bornhauser, M.; Von Bonin, M.; Wermke, M.; Feldmann, A.; Bachmann, M.F.; 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]
- Vergez, F.; Green, A.S.; Tamburini, J.; Sarry, J.-E.; Gaillard, B.; Cornillet-Lefebvre, P.; Pannetier, M.; Neyret, A.; Chapuis, N.; Ifrah, N.; et al. High levels of CD34+CD38low/−CD123+ blasts are predictive of an adverse outcome in acute myeloid leukemia: A Groupe Ouest-Est des Leucémies Aiguës et Maladies du Sang (GOELAMS) study. Haematologica 2011, 96, 1792–1798. [Google Scholar] [CrossRef]
- Angelini, D.F.; Ottone, T.; Guerrera, G.; Lavorgna, S.; Cittadini, M.; Buccisano, F.; De Bardi, M.; Gargano, F.; Maurillo, L.; Divona, M.; et al. A Leukemia-Associated CD34/CD123/CD25/CD99+ Immunophenotype Identifies FLT3-Mutated Clones in Acute Myeloid Leukemia. Clin. Cancer Res. 2015, 21, 3977–3985. [Google Scholar] [CrossRef] [Green Version]
- Yan, B.; Chen, Q.; Shimada, K.; Tang, M.; Li, H.; Gurumurthy, A.; Khoury, J.D.; Xu, B.; Huang, S.; Qiu, Y. Histone deacetylase inhibitor targets CD123/CD47-positive cells and reverse chemoresistance phenotype in acute myeloid leukemia. Leukemia 2019, 33, 931–944. [Google Scholar] [CrossRef]
- Al-Hussaini, M.; Rettig, M.P.; Ritchey, J.K.; Karpova, D.; Uy, G.L.; Eissenberg, L.G.; Gao, F.; Eades, W.C.; Bonvini, E.; Chichili, G.R.; et al. Targeting CD123 in acute myeloid leukemia using a T-cell–directed dual-affinity retargeting platform. Blood 2016, 127, 122–131. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Pizzitola, I.; Anjosafonso, F.; Rouaultpierre, K.; Lassailly, F.; Tettamanti, S.; Spinelli, O.; Biondi, A.; Biagi, E.; Bonnet, D. Chimeric antigen receptors against CD33/CD123 antigens efficiently target primary acute myeloid leukemia cells in vivo. Leukemia 2014, 28, 1596–1605. [Google Scholar] [CrossRef] [PubMed]
- Bonifant, C.L.; Szoor, A.; Torres, D.; Joseph, N.; Velasquez, M.P.; Iwahori, K.; Gaikwad, A.; Nguyen, P.; Arber, C.; Song, X.-T.; et al. CD123-Engager T Cells as a Novel Immunotherapeutic for Acute Myeloid Leukemia. Mol. Ther. 2016, 24, 1615–1626. [Google Scholar] [CrossRef] [Green Version]
- Xie, L.H.; Biondo, M.; Busfield, S.J.; Arruda, A.; Yang, X.; Vairo, G.; Minden, M.D. CD123 target validation and preclinical evaluation of ADCC activity of anti-CD123 antibody CSL362 in combination with NKs from AML patients in remission. Blood Cancer J. 2017, 7, e567. [Google Scholar] [CrossRef] [PubMed]
- Han, L.; Jorgensen, J.L.; Brooks, C.; Shi, C.; Zhang, Q.; González, G.M.N.; Cavazos, A.; Pan, R.; Mu, H.; Wang, S.A.; et al. Antileukemia Efficacy and Mechanisms of Action of SL-101, a Novel Anti-CD123 Antibody Conjugate, in Acute Myeloid Leukemia. Clin. Cancer Res. 2017, 23, 3385–3395. [Google Scholar] [CrossRef] [Green Version]
- Kovtun, Y.; Jones, G.E.; Adams, S.; Harvey, L.; Audette, C.A.; Wilhelm, A.; Bai, C.; Rui, L.; Laleau, R.; Liu, F.; et al. A CD123-targeting antibody-drug conjugate, IMGN632, designed to eradicate AML while sparing normal bone marrow cells. Blood Adv. 2018, 2, 848–858. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Taussig, D.C. 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]
- De Propris, M.S.; Raponi, S.; Diverio, D.; Milani, M.L.; Meloni, G.; Falini, B.; Foa, R.; Guarini, A. High CD33 expression levels in acute myeloid leukemia cells carrying the nucleophosmin (NPM1) mutation. Haematologica 2011, 96, 1548–1551. [Google Scholar] [CrossRef] [Green Version]
- Khan, N.; on behalf of the UK NCRI-AML Study Group; Hills, R.K.; Virgo, P.; Couzens, S.; Clark, N.; Gilkes, A.; Richardson, P.; Knapper, S.; Grimwade, D.; et al. Expression of CD33 is a predictive factor for effect of gemtuzumab ozogamicin at different doses in adult acute myeloid leukaemia. Leukemia 2017, 31, 1059–1068. [Google Scholar] [CrossRef] [PubMed] [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]
- Pollard, J.A.; Alonzo, T.A.; Loken, M.; Gerbing, R.B.; Ho, P.A.; Bernstein, I.D.; Raimondi, S.C.; Hirsch, B.; Franklin, J.; Walter, R.B.; et al. Correlation of CD33 expression level with disease characteristics and response to gemtuzumab ozogamicin containing chemotherapy in childhood AML. Blood 2012, 119, 3705–3711. [Google Scholar] [CrossRef] [Green Version]
- Friedrich, M.; Henn, A.; Raum, T.; Bajtus, M.; Matthes, K.; Hendrich, L.; Wahl, J.; Hoffmann, P.; Kischel, R.; Kvesic, M.; et al. Preclinical Characterization of AMG 330, a CD3/CD33-Bispecific T-Cell–Engaging Antibody with Potential for Treatment of Acute Myelogenous Leukemia. Mol. Cancer Ther. 2014, 13, 1549–1557. [Google Scholar] [CrossRef] [Green Version]
- Herrmann, M.; Krupka, C.; Deiser, K.; Brauchle, B.; Marcinek, A.; Wagner, A.O.; 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, 2484–2494. [Google Scholar] [CrossRef] [Green Version]
- Neumann, K.; Castineiras-Vilarino, M.; Höckendorf, U.; Hannesschläger, N.; Lemeer, S.; Kupka, D.; Meyermann, S.; Lech, M.; Anders, H.-J.; Kuster, B.; et al. Clec12a Is an Inhibitory Receptor for Uric Acid Crystals that Regulates Inflammation in Response to Cell Death. Immunity 2014, 40, 389–399. [Google Scholar] [CrossRef] [Green Version]
- Redelinghuys, P.; Whitehead, L.; Augello, A.; Drummond, R.A.; Levesque, J.-M.; Vautier, S.; Reid, D.M.; Kerscher, B.; Taylor, J.A.; Nigrovic, P.A.; et al. MICL controls inflammation in rheumatoid arthritis. Ann. Rheum. Dis. 2015, 75, 1386–1391. [Google Scholar] [CrossRef] [Green Version]
- Van Rhenen, A.; Van Dongen, G.A.M.S.; Kelder, A.; Rombouts, E.J.; Feller, N.; Moshaver, B.; Walsum, M.S.-V.; Zweegman, S.; Ossenkoppele, G.J.; Schuurhuis, G.J. 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]
- Hutten, T.J.A.; Thordardottir, S.; Fredrix, H.; Janssen, L.; Woestenenk, R.; Tel, J.; Joosten, B.; Cambi, A.; Heemskerk, M.H.M.; Franssen, G.M.; et al. CLEC12A-Mediated Antigen Uptake and Cross-Presentation by Human Dendritic Cell Subsets Efficiently Boost Tumor-Reactive T Cell Responses. J. Immunol. 2016, 197, 2715–2725. [Google Scholar] [CrossRef]
- Zheng, B.; Yu, S.-F.; Del Rosario, G.; Leong, S.R.; Lee, G.Y.; Vij, R.; Chiu, C.P.; 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]
- 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] [Green Version]
- Ma, H.; Padmanabhan, I.S.; Parmar, S.; Gong, Y. Targeting CLL-1 for acute myeloid leukemia therapy. J. Hematol. Oncol. 2019, 12, 1–11. [Google Scholar] [CrossRef]
- Zhao, X.; Singh, S.; Pardoux, C.; Zhao, J.; Hsi, E.D.; Abo, A.; Korver, W. Targeting C-type lectin-like molecule-1 for antibody-mediated immunotherapy in acute myeloid leukemia. Haematologica 2009, 95, 71–78. [Google Scholar] [CrossRef]
- 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]
- 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. 2014, 53, 9841–9845. [Google Scholar] [CrossRef] [Green Version]
- Van Loo, P.F.; Doornbos, R.; Dolstra, H.; Shamsili, S.; Bakker, L. Preclinical Evaluation of MCLA117, a CLEC12AxCD3 Bispecific Antibody Efficiently Targeting a Novel Leukemic Stem Cell Associated Antigen in AML. Blood 2015, 126, 325. [Google Scholar] [CrossRef]
- Tashiro, H.; Sauer, T.; Shum, T.; Parikh, K.; Mamonkin, M.; Omer, B.; Rouce, R.H.; Lulla, P.; Rooney, C.M.; Gottschalk, S.; et al. Treatment of Acute Myeloid Leukemia with T Cells Expressing Chimeric Antigen Receptors Directed to C-type Lectin-like Molecule 1. Mol. Ther. 2017, 25, 2202–2213. [Google Scholar] [CrossRef] [Green Version]
- Laborda, E.; Mazagova, M.; Shao, S.; Wang, X.; Quirino, H.; Woods, A.K.; Hampton, E.N.; Rodgers, D.T.; Kim, C.H.; Schultz, P.G.; et al. Development of A Chimeric Antigen Receptor Targeting C-Type Lectin-Like Molecule-1 for Human Acute Myeloid Leukemia. Int. J. Mol. Sci. 2017, 18, 2259. [Google Scholar] [CrossRef] [Green Version]
- Kenderian, S.S.; Ruella, M.; Shestova, O.; Klichinsky, M.; Kim, M.Y.; Soderquist, C.; Bagg, A.; Singh, R.; Richardson, C.; Young, R.M.; et al. Leukemia Stem Cells Are Characterized By CLEC12A Expression and Chemotherapy Refractoriness That Can be Overcome By Targeting with Chimeric Antigen Receptor T Cells. Blood 2016, 128, 766. [Google Scholar] [CrossRef]
- 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] [Green Version]
- De Togni, E.; Kim, M.Y.; Cooper, M.L.; Ritchey, J.; O’Neal, J.; Niswonger, J.; DiPersio, J.F. Chimeric Antigen Receptor T Cells Specific for CLL-1 for Treatment of Acute Myeloid Leukemia. Blood 2018, 132, 2205. [Google Scholar] [CrossRef]
- Arvindam, U.S.; Van Hauten, P.; Hallstrom, C.; Vallera, D.A.; Dolstra, H.; Miller, J.S.; Felices, M. CD16-IL15-CLEC12A Trispecific Killer Engager (TriKE) Drives NK Cell Expansion, Activation, and Antigen Specific Killing of Cancer Stem Cells in Acute Myeloid Leukemia. Blood 2018, 132, 1454. [Google Scholar] [CrossRef]
- Gao, X.; Lin, J.; Gao, L.; Deng, A.; Lu, X.; Li, Y.; Wang, L.; Yu, L. High Expression of c-kit mRNA Predicts Unfavorable Outcome in Adult Patients with t(8;21) Acute Myeloid Leukemia. PLoS ONE 2015, 10, e0124241. [Google Scholar] [CrossRef] [Green Version]
- Myburgh, R.; Kiefer, J.D.; Russkamp, N.F.; Magnani, C.F.; Nuñez, N.; Simonis, A.; Pfister, S.; Wilk, C.M.; McHugh, D.; Friemel, J.; et al. Anti-human CD117 CAR T-cells efficiently eliminate healthy and malignant CD117-expressing hematopoietic cells. Leukemia 2020, 34, 2688–2703. [Google Scholar] [CrossRef]
- Kikushige, Y.; Yoshimoto, G.; Miyamoto, T.; Iino, T.; Mori, Y.; Iwasaki, H.; Niiro, H.; Takenaka, K.; Nagafuji, K.; Harada, M.; et al. Human Flt3 Is Expressed at the Hematopoietic Stem Cell and the Granulocyte/Macrophage Progenitor Stages to Maintain Cell Survival. J. Immunol. 2008, 180, 7358–7367. [Google Scholar] [CrossRef] [Green Version]
- Stirewalt, D.L.; Radich, J.P. The role of FLT3 in haematopoietic malignancies. Nat. Rev. Cancer 2003, 3, 650–665. [Google Scholar] [CrossRef]
- Perl, A.E.; Martinelli, G.; Cortes, J.E.; Neubauer, A.; Berman, E.; Paolini, S.; Montesinos, P.; Baer, M.R.; Larson, R.A.; Ustun, C.; et al. Gilteritinib or Chemotherapy for Relapsed or Refractory FLT3-Mutated AML. N. Engl. J. Med. 2019, 381, 1728–1740. [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]
- Cortes, J.E.; Khaled, S.; Martinelli, G.; Perl, A.; Ganguly, S.; Russell, N.; Krämer, A.; Dombret, H.; Hogge, D.; Jonas, B.; et al. Quizartinib versus salvage chemotherapy in relapsed or refractory FLT3-ITD acute myeloid leukaemia (QuANTUM-R): A multicentre, randomised, controlled, open-label, phase 3 trial. Lancet Oncol. 2019, 20, 984–997. [Google Scholar] [CrossRef]
- Jetani, H.; Garcia-Cadenas, I.; Nerreter, T.; Thomas, S.; Rydzek, J.; Meijide, J.B.; Bonig, H.; Herr, W.; Sierra, J.; Einsele, H.; et al. CAR T-cells targeting FLT3 have potent activity against FLT3−ITD+ AML and act synergistically with the FLT3-inhibitor crenolanib. Leukemia 2018, 32, 1168–1179. [Google Scholar] [CrossRef]
- Seitz, C.M.; Kieble, C.V.; Illi, C.C.; Reiter, C.S.; Grote, M.S.; Mittelstaet, J.; Lock, M.D.; Kaiser, A.; Schleicher, S.; Handgretinger, R.; et al. Combinatorial Targeting of Multiple Shared Antigens By Adapter-CAR-T Cells (aCAR-Ts) Allows Target Cell Discrimination and Specific Lysis Based on Differential Expression Profiles. Blood 2018, 132, 4543. [Google Scholar] [CrossRef]
- Daver, N.; Basu, S.; Garcia-Manero, G.; Cortes, J.E.; Ravandi, F.; Jabbour, E.J.; Hendrickson, R.S.; Pierce, B.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]
- Daver, N.G.; Garcia-Manero, G.; Basu, S.; Cortes, J.E.; Ravandi, F.; Kadia, T.M.; Konopleva, M.Y.; Jabbour, E.J.; Dinardo, C.D.; Assi, R.; et al. Safety, Efficacy, and Biomarkers of Response to Azacitidine (AZA) with Nivolumab (Nivo) and AZA with Nivo and Ipilimumab (Ipi) in Relapsed/Refractory Acute Myeloid Leukemia: A Non-Randomized, Phase 2 Study. Blood 2018, 132, 906. [Google Scholar] [CrossRef]
- Ravandi, F.; Assi, R.; Daver, N.; Benton, C.B.; Kadia, T.; Thompson, P.; Borthakur, G.; Alvarado, Y.; Jabbour, E.J.; Konopleva, M.; et al. Idarubicin, cytarabine, and nivolumab in patients with newly diagnosed acute myeloid leukaemia or high-risk myelodysplastic syndrome: A single-arm, phase 2 study. Lancet Haematol. 2019, 6, e480–e488. [Google Scholar] [CrossRef]
- Lindblad, E.K.; Thompson, J.; Gui, G.; Valdez, J.; Worthy, T.; Tekleab, H.; Hughes, T.; Goswami, M.; Oetjen, K.; Kim, N.-Y.; et al. Pembrolizumab and Decitabine for Refractory or Relapsed Acute Myeloid Leukemia. Blood 2018, 132, 1437. [Google Scholar] [CrossRef]
- 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]
- 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]
- Zeidan, A.M.; Knaus, H.A.; Robinson, T.M.; Towlerton, A.M.; Warren, E.H.; Zeidner, J.F.; Blackford, A.L.; Duffield, A.S.; Rizzieri, D.; Frattini, M.G.; et al. A Multi-center Phase I Trial of Ipilimumab in Patients with Myelodysplastic Syndromes following Hypomethylating Agent Failure. Clin. Cancer Res. 2018, 24, 3519–3527. [Google Scholar] [CrossRef] [Green Version]
- Borate, U.; Esteve, J.; Porkka, K.; Knapper, S.; Vey, N.; Scholl, S.; Garcia-Manero, G.; Wermke, M.; Janssen, J.; Traer, E.; et al. Phase Ib Study of the Anti-TIM-3 Antibody MBG453 in Combination with Decitabine in Patients with High-Risk Myelodysplastic Syndrome (MDS) and Acute Myeloid Leukemia (AML). Blood 2019, 134, 570. [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] [Green Version]
- 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] [Green Version]
- Tang, X.; Yang, L.; Li, Z.; Nalin, A.P.; Dai, H.; Xu, T.; Yin, J.; You, F.; Zhu, M.; Shen, W.; et al. First-in-man clinical trial of CAR NK-92 cells: Safety test of CD33-CAR NK-92 cells in patients with relapsed and refractory acute myeloid leukemia. Am. J. Cancer Res. 2018, 8, 1899. [Google Scholar]
- Suh, H.C.; Pohl, K.A.; Termini, C.; Kan, J.; Timmerman, J.M.; Slamon, D.J.; Chute, J.P. Bioengineered Autologous Dendritic Cells Enhance CAR T Cell Cytotoxicity By Providing Cytokine Stimulation and Intratumoral Dendritic Cells. Blood 2018, 132, 3693. [Google Scholar] [CrossRef]
- 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]
- Sallman, D.; Kerre, T.; Poire, X.; Havelange, V.; Lewalle, P.; Davila, M.L.; Wang, E.S.; Dekker, D.; Snykers, S.; Sotiropoulou, P.A.; et al. Remissions in Relapse/Refractory Acute Myeloid Leukemia Patients Following Treatment with NKG2D CAR-T Therapy without a Prior Preconditioning Chemotherapy. Blood 2018, 132, 902. [Google Scholar] [CrossRef]
- Velasquez, M.P.; Bonifant, C.L.; Gottschalk, S. Redirecting T cells to hematological malignancies with bispecific antibodies. Blood 2018, 131, 30–38. [Google Scholar] [CrossRef]
- Ravandi, F.; Stein, A.S.; Kantarjian, H.M.; Walter, P.M.R.B.; Paschka, P.; Jongen-Lavrencic, M.; Ossenkoppele, G.J.; Yang, Z.; Mehta, B.; Subklewe, M. A Phase 1 First-in-Human Study of AMG 330, an Anti-CD33 Bispecific T-Cell Engager (BiTE®) Antibody Construct, in Relapsed/Refractory Acute Myeloid Leukemia (R/R AML). Blood 2018, 132, 25. [Google Scholar] [CrossRef]
- Westervelt, P.; Roboz, G.J.; Cortes, J.E.; Kantarjian, H.M.; Lee, S.; Rettig, M.P.; Han, T.H.; Guenot, J.; Feldman, E.J.; DiPersio, J.F. Phase 1 First-in-Human Trial of AMV564, a Bivalent Bispecific (2x2) CD33/CD3 T-Cell Engager, in Patients with Relapsed/Refractory Acute Myeloid Leukemia (AML). Blood 2018, 132, 1455. [Google Scholar] [CrossRef]
- Ravandi, F.; Bashey, A.; Foran, J.M.; Stock, W.; Mawad, R.; Blum, W.; Saville, M.W.; Johnson, B.C.M.; Vanasse, K.G.J.; Ly, T.; et al. Complete Responses in Relapsed/Refractory Acute Myeloid Leukemia (AML) Patients on a Weekly Dosing Schedule of XmAb14045, a CD123 x CD3 T Cell-Engaging Bispecific Antibody: Initial Results of a Phase 1 Study. Blood 2018, 132, 763. [Google Scholar] [CrossRef]
- Uy, G.L.; Rettig, M.P.; Vey, N.; Godwin, J.; Foster, M.C.; Rizzieri, D.A.; Arellano, M.L.; Topp, M.S.; Huls, G.; Jongen-Lavrencic, M.; et al. Phase 1 Cohort Expansion of Flotetuzumab, a CD123×CD3 Bispecific Dart® Protein in Patients with Relapsed/Refractory Acute Myeloid Leukemia (AML). Blood 2018, 132, 764. [Google Scholar] [CrossRef]
- Wei, A.H.; Fong, C.Y.; Montesinos, P.; Calbacho, M.; Gil, J.S.; De Oteyza, J.P.; Rowe, J.M.; Wolach, O.; Ofran, Y.; Moshe, Y.; et al. A Phase 1 Study of Flotetuzumab, a CD123 x CD3 DART® Protein, Combined with MGA012, an Anti-PD-1 Antibody, in Patients with Relapsed or Refractory Acute Myeloid Leukemia. Blood 2019, 134, 2662. [Google Scholar] [CrossRef]
- Amadori, S.; Suciu, S.; Selleslag, D.; Aversa, F.; Gaidano, G.; Musso, M.; Annino, L.; Venditti, A.; Voso, M.T.; Mazzone, C.; et al. Gemtuzumab Ozogamicin Versus Best Supportive Care in Older Patients With Newly Diagnosed Acute Myeloid Leukemia Unsuitable for Intensive Chemotherapy: Results of the Randomized Phase III EORTC-GIMEMA AML-19 Trial. J. Clin. Oncol. 2016, 34, 972–979. [Google Scholar] [CrossRef] [PubMed]
- Heinhuis, K.; Ros, W.; Kok, M.; Steeghs, N.; Beijnen, J.; Schellens, J. Enhancing antitumor response by combining immune checkpoint inhibitors with chemotherapy in solid tumors. Ann. Oncol. 2019, 30, 219–235. [Google Scholar] [CrossRef] [PubMed]
- Dagoglu, N.; Karaman, S.; Caglar, H.B.; Oral, E.N. Abscopal Effect of Radiotherapy in the Immunotherapy Era: Systematic Review of Reported Cases. Cureus 2019, 11. [Google Scholar] [CrossRef] [Green Version]
Author | Object(s) | Disease State | Agent(s) | Dosing | Phase | Response Rate | Median Survival | |
---|---|---|---|---|---|---|---|---|
Daver, et al., 2016 | AML | Relapsed after prior therapy | Nivolumab | 3 mg/kg on Day 1, 14 (every 4–5 weeks) | Ib/II | CR/CRi HI | 18% (6/51) 15% (5/51) | 9.3 mo. [1.8–14.3] |
+ Azacitidine | 75 mg/m2 on Day 1–7 (every 4–5 weeks) | |||||||
Daver, et al., 2018 | AML | Relapsed or refractory | Nivolumab + Azacitidin | [Cohort 1] Not Reported | II | CR/CRi HI Prolonged SD | 21% (15/70) 10% (7/70) 9% (6/70) | 16.1 mo. |
Nivolumab+ Azacitidin+ Ipilimimab | [Cohort 2] Not Reported | II | CR/CRi Prolonged SD | 36% (6/20) 10% (2/20) | Not Reached (1-yr. OS 58%) | |||
Ravandi, et al., 2019 | AML and high-risk MDS | Newly diagnosed | Idarubicin + Cytarabine + Nivolumab | 12 mg/m2 on Day 1–3 1.5 g/m2 on Day 1–4 3 mg/kg (every 2 weeks) *started on Day 24 | II | CR/CRi Negative MRD | 78% (34/44) 41% (18/34) | 18.5 mo. [10.8–28.8] |
Davids, et al., 2016 | Hematologic malignancies (AML, HL, NHL, MDS, MM, MPN, ALL) | Relapsed after allo-HSCT | Ipilimumab | 3 or 10 mg/kg (every 3 weeks for 4 doses then every 12 weeks for upto 6 doses) *All reseposive cases recieved 10 mg/kg. | I/Ib | CR PR | 23% (5/28) 9% (2/22) | Not Reported |
Bashey, et al., 2009 | Malignancies (AML, HL, NHL, MM, CML, CLL, Breast cancer, RCC) | Relapsed after allo-HSCT | Ipilimumab | 0.1 to 3.0 mg/kg (every 60 days) *Dose-escalating model. | I | CR PR | 6.9% (2/29) 3.4% (1/29) | 24.7 mo. |
Zeidan, et al., 2018 | MDS | Refractory to HMAs | Ipilimumab | 3 or 10 mg/kg (every 3 weeks for 4 doses then every 12 weeks for upto 8 doses) | I | Marrow CR Prolonged SD | 3.4% (1/29) 24% (7/29) | Not Reported |
Lindblad, et al., 2018 | AML | Relapsed or refractory | Pembrolizumab + Decitabine | 200 mg/body (every 3 weeks) 20 mg/m2 on Day 8–12, 15–19 (every 6 weeks) | I/II | CR SD | 10% (1/10) 40% (4/10) | 7 mo. [2,3,4,5,6,7,8,9,10,11,12,13,14] |
Borate, et al., 2019 | AML and high-risk MDS | Ineligible to standard therapy | MBG453 (anti-TIM-3) + Decitabine | Escalating dose from 240 to 800 mg/body (every 2 weeks or 4 weeks) 20 mg/m2 on Day 1–5 (every 4 weeks) | Ib | CR/CRi PR Blasts halved | 23% (7/31) 6% (2/31)2 6% (8/31) | Exposure durations 2.1–17.9 months |
Authors | Objects | Disease State | Agents | Target(s) | Phase | Clinical Outcome |
---|---|---|---|---|---|---|
Rithchie, et al., 2013 | AML | Rel./ref. | CAR-T (2nd gen.) | Lewis-Y antigen | I | Transient decrease of blasts in 1 of 4 patients 14–38% of Transduction efficiency |
Wang, et al., 2015 | AML | Rel./ref. | CAR-T (2nd gen.) | CD33 | I | Marked reduction of marrow blasts for 9 weeks in 1 patient |
Sallman, et al., 2018 | Solid tumors and hematologic malignancies | Rel./ref. | CAR-T | NKG2D | I | 1 CRh and 2 CRi of 7 patients with AML |
Liu, et al., 2018 | JMML | Rel./ref. | Compound CAR-T | CLL-1 and CD33 | I | CR with negative MRD in 1 patient |
Tang, et al., 2018 | AML | Rel./ref. | CAR-NK | CD33 | I | Decrease of MRD and WT-1 in 1 of 3 patients |
Author | Object | Agent(s) | Class | Targets | Phase | Clinical Outcome | |
---|---|---|---|---|---|---|---|
Ravandi, et al., 2018 | RR-AML | AMG330 | BiTE | CD33 and CD3 | I | CR/CRi CRS | 11.4% (4 of 35) 42.1% (15 of 35) |
Eissenberg, et al., 2018 | RR-AML | AMV564 | BiTE | CD33 and CD3 | I | CR/CRi CRS | 66.7% (12 of 18) 5.7% (1 of 18) |
Ravandi, et al., 2018 | RR-AML | XmAb14045 | BiTE | CD123 and CD3 | I | CR/CRi CRS | 23.1% (3 of 13) 77.9% (49 of 63) |
Uy, et al., 2018 | RR-AML | Flotetuzumab | DART | CD123 and CD3 | I/II | CR/CRi Severe CRS | 18.5% (5 of 27) 13.4% (4 of 30) |
Wei, et al., 2019 | RR-AML | Flotetuzumab +MGA012 | DART+ ICI | CD123 and CD3 plus PD-1 | I | Not reported |
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
Tabata, R.; Chi, S.; Yuda, J.; Minami, Y. Emerging Immunotherapy for Acute Myeloid Leukemia. Int. J. Mol. Sci. 2021, 22, 1944. https://doi.org/10.3390/ijms22041944
Tabata R, Chi S, Yuda J, Minami Y. Emerging Immunotherapy for Acute Myeloid Leukemia. International Journal of Molecular Sciences. 2021; 22(4):1944. https://doi.org/10.3390/ijms22041944
Chicago/Turabian StyleTabata, Rikako, SungGi Chi, Junichiro Yuda, and Yosuke Minami. 2021. "Emerging Immunotherapy for Acute Myeloid Leukemia" International Journal of Molecular Sciences 22, no. 4: 1944. https://doi.org/10.3390/ijms22041944
APA StyleTabata, R., Chi, S., Yuda, J., & Minami, Y. (2021). Emerging Immunotherapy for Acute Myeloid Leukemia. International Journal of Molecular Sciences, 22(4), 1944. https://doi.org/10.3390/ijms22041944