Skip to main content
Top

25-07-2017 | Acute myeloid leukemia | Article

Recent developments in immunotherapy of acute myeloid leukemia

Journal: Journal of Hematology & Oncology

Authors: Felix S. Lichtenegger, Christina Krupka, Sascha Haubner, Thomas Köhnke, Marion Subklewe

Publisher: BioMed Central

Abstract

The advent of new immunotherapeutic agents in clinical practice has revolutionized cancer treatment in the past decade, both in oncology and hematology. The transfer of the immunotherapeutic concepts to the treatment of acute myeloid leukemia (AML) is hampered by various characteristics of the disease, including non-leukemia-restricted target antigen expression profile, low endogenous immune responses, and intrinsic resistance mechanisms of the leukemic blasts against immune responses. However, considerable progress has been made in this field in the past few years.
Within this manuscript, we review the recent developments and the current status of the five currently most prominent immunotherapeutic concepts: (1) antibody-drug conjugates, (2) T cell-recruiting antibody constructs, (3) chimeric antigen receptor (CAR) T cells, (4) checkpoint inhibitors, and (5) dendritic cell vaccination. We focus on the clinical data that has been published so far, both for newly diagnosed and refractory/relapsed AML, but omitting immunotherapeutic concepts in conjunction with hematopoietic stem cell transplantation. Besides, we have included important clinical trials that are currently running or have recently been completed but are still lacking full publication of their results.
While each of the concepts has its particular merits and inherent problems, the field of immunotherapy of AML seems to have taken some significant steps forward. Results of currently running trials will reveal the direction of further development including approaches combining two or more of these concepts.
Literature
1.
Lichtenegger FS, Schnorfeil FM, Hiddemann W, Subklewe M. Current strategies in immunotherapy for acute myeloid leukemia. Immunotherapy. 2013;5(1):63–78.CrossRefPubMed
2.
Lichtenegger FS, Krupka C, Köhnke T, Subklewe M. Immunotherapy for acute myeloid leukemia. Semin Hematol. 2015;52(3):207–14.CrossRefPubMed
3.
Cornelissen JJ, Gratwohl A, Schlenk RF, Sierra J, Bornhäuser M, Juliusson G, et al. The European LeukemiaNet AML Working Party consensus statement on allogeneic HSCT for patients with AML in remission: an integrated-risk adapted approach. Nat Rev Clin Oncol. 2012;9(10):579–90.CrossRefPubMed
4.
Stelljes M, Krug U, Beelen DW, Braess J, Sauerland MC, Heinecke A, et al. Allogeneic transplantation versus chemotherapy as postremission therapy for acute myeloid leukemia: a prospective matched pairs analysis. J Clin Oncol. 2014;32(4):288–96.CrossRefPubMed
5.
Ramos NR, Mo CC, Karp JE, Hourigan CS. Current approaches in the treatment of relapsed and refractory acute myeloid leukemia. J Clin Med. 2015;4(4):665–95.CrossRefPubMedPubMedCentral
6.
Thomas DA, O'Brien S, Kantarjian HM. Monoclonal antibody therapy with rituximab for acute lymphoblastic leukemia. Hematol Oncol Clin North Am. 2009;23(5):949–71. v.CrossRefPubMedPubMedCentral
7.
Kantarjian HM, DeAngelo DJ, Stelljes M, Martinelli G, Liedtke M, Stock W, et al. Inotuzumab ozogamicin versus standard therapy for acute lymphoblastic leukemia. N Engl J Med. 2016;375(8):740–53.CrossRefPubMed
8.
Aldoss I, Bargou RC, Nagorsen D, Friberg GR, Baeuerle PA, Forman SJ. Redirecting T cells to eradicate B-cell acute lymphoblastic leukemia: bispecific T-cell engagers and chimeric antigen receptors. Leukemia. 2017.
9.
Topp MS, Gökbuget N, Stein AS, Zugmaier G, O'Brien S, Bargou RC, et al. Safety and activity of blinatumomab for adult patients with relapsed or refractory B-precursor acute lymphoblastic leukaemia: a multicentre, single-arm, phase 2 study. Lancet Oncol. 2015;16(1):57–66.CrossRefPubMed
10.
Holzinger A, Barden M, Abken H. The growing world of CAR T cell trials: a systematic review. Cancer Immunol Immunother. 2016;65(12):1433–50.CrossRefPubMed
11.
Rotiroti MC, Arcangeli S, Casucci M, Perriello V, Bondanza A, Biondi A, et al. Acute myeloid leukemia targeting by chimeric antigen receptor T cells: bridging the gap from preclinical modeling to human studies. Hum Gene Ther. 2016.
12.
Hoseini SS, Cheung NK. Acute myeloid leukemia targets for bispecific antibodies. Blood Cancer J. 2017;7(2), e522.CrossRefPubMedPubMedCentral
13.
Hude I, Sasse S, Engert A, Bröckelmann PJ. The emerging role of immune checkpoint inhibition in malignant lymphoma. Haematologica. 2017;102(1):30–42.CrossRefPubMedPubMedCentral
14.
Peters C, Brown S. Antibody-drug conjugates as novel anti-cancer chemotherapeutics. Biosci Rep. 2015;35(4), e00225.CrossRefPubMedPubMedCentral
15.
Bross PF, Beitz J, Chen G, Chen XH, Duffy E, Kieffer L, et al. Approval summary: gemtuzumab ozogamicin in relapsed acute myeloid leukemia. Clin Cancer Res. 2001;7(6):1490–6.PubMed
16.
Petersdorf H, Kopecky J, Slovak M, Willman C, Nevill T, Brandwein J, et al. A phase III study of gemtuzumab ozogamicin during induction and post-consolidation therapy in younger patients with acute myeloid leukemia. Blood. 2013;121(24):4854–60.CrossRefPubMedPubMedCentral
17.
Hills RK, Castaigne S, Appelbaum FR, Delaunay J, Petersdorf S, Othus M, 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(9):986–96.CrossRefPubMedPubMedCentral
18.
Loke J, Khan JN, Wilson JS, Craddock C, Wheatley K. Mylotarg has potent anti-leukaemic effect: a systematic review and meta-analysis of anti-CD33 antibody treatment in acute myeloid leukaemia. Ann Hematol. 2015;94(3):361–73.CrossRefPubMed
19.
Amadori S, Suciu S, Selleslag D, Aversa F, Gaidano G, Musso M, 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(9):972–9.CrossRefPubMed
20.
Nand S, Othus M, Godwin JE, Willman CL, Norwood TH, Howard DS, et al. A phase 2 trial of azacitidine and gemtuzumab ozogamicin therapy in older patients with acute myeloid leukemia. Blood. 2013;122(20):3432–9.CrossRefPubMedPubMedCentral
21.
Walter RB, Medeiros BC, Gardner KM, Orlowski KF, Gallegos L, Scott BL, et al. Gemtuzumab ozogamicin in combination with vorinostat and azacitidine in older patients with relapsed or refractory acute myeloid leukemia: a phase I/II study. Haematologica. 2014;99(1):54–9.CrossRefPubMedPubMedCentral
22.
Daver N, Kantarjian H, Ravandi F, Estey E, Wang X, Garcia-Manero G, et al. A phase II study of decitabine and gemtuzumab ozogamicin in newly diagnosed and relapsed acute myeloid leukemia and high-risk myelodysplastic syndrome. Leukemia. 2016;30(2):268–73.CrossRefPubMed
23.
Nguyen DH, Ball ED, Varki A. Myeloid precursors and acute myeloid leukemia cells express multiple CD33-related Siglecs. Exp Hematol. 2006;34(6):728–35.CrossRefPubMed
24.
Ehninger A, Kramer M, Röllig C, Thiede C, Bornhäuser M, von Bonin M, et al. Distribution and levels of cell surface expression of CD33 and CD123 in acute myeloid leukemia. Blood Cancer J. 2014;4, e218.CrossRefPubMedPubMedCentral
25.
Krupka C, Kufer P, Kischel R, Zugmaier G, Bögeholz J, Köhnke T, et al. CD33 target validation and sustained depletion of AML blasts in long-term cultures by the bispecific T-cell-engaging antibody AMG 330. Blood. 2014;123(3):356–65.CrossRefPubMed
26.
McCombs JR, Owen SC. Antibody drug conjugates: design and selection of linker, payload and conjugation chemistry. AAPS J. 2015;17(2):339–51.CrossRefPubMedPubMedCentral
27.
Linenberger ML. CD33-directed therapy with gemtuzumab ozogamicin in acute myeloid leukemia: progress in understanding cytotoxicity and potential mechanisms of drug resistance. Leukemia. 2005;19(2):176–82.CrossRefPubMed
28.
Kung Sutherland MS, Walter RB, Jeffrey SC, Burke PJ, Yu C, Kostner H, et al. SGN-CD33A: a novel CD33-targeting antibody-drug conjugate using a pyrrolobenzodiazepine dimer is active in models of drug-resistant AML. Blood. 2013;122(8):1455–63.CrossRefPubMed
29.
Bixby DL, Stein AS, Fathi AT, Kovavsovics TJ, Levy MY, Erba HP, et al. Vadastuximab talirine monotherapy in older patients with treatment naive CD33-positive acute myeloid leukemia (AML). Blood. 2016;128:590.
30.
Fathi AT, Erba HP, Lancet JE, Stein EM, Ravandi F, Faderl S, et al. Vadastuximab talirine plus hypomethylating agents: a well-tolerated regimen with high remission rate in frontline older patients with acute myeloid leukemia (AML). Blood. 2016;128:591.
31.
Erba HP, Levy MY, Vasu S, Stein AS, Fathi AT, Maris MB, et al. A phase 1b study of vadastuximab talirine in combination with 7 + 3 induction therapy for patients with newly diagnosed acute myeloid leukemia (AML). Blood. 2016;128:211.
32.
Seattle Genetics, Inc. Seattle Genetics announces clinical hold on several phase 1 trials of vadastuximab talirine (SGN-CD33A). http://​www.​businesswire.​com/​news/​home/​20161227005087/​en/​Seattle-Genetics-Announces-Clinical-Hold-Phase-1. Accessed 6 Mar 2017.
33.
Testa U, Pelosi E, Frankel A. CD 123 is a membrane biomarker and a therapeutic target in hematologic malignancies. Biomark Res. 2014;2(1):4.CrossRefPubMedPubMedCentral
34.
Krystal WM, Walker R, Fishkin N, Audette C, Kovtun Y, Romanelli A. IMGN779, a CD33-targeted antibody-drug conjugate (ADC) with a novel DNA-alkylating effector molecule, induces DNA damage, cell cycle arrest, and apoptosis in AML cells. Blood. 2015;126:1366.
35.
Whiteman KR, Noordhuis P, Walker R, Watkins K, Kovtun Y, Harvey L, et al. The antibody-drug conjugate (ADC) IMGN779 is highly active in vitro and in vivo against acute myeloid leukemia (AML) with FLT3-ITD mutations. Blood. 2014;124:2321.
36.
Portwood S, Puchalski RA, Walker RM, Wang ES. Combining IMGN779, a novel anti-CD33 antibody-drug conjugate (ADC), with the PARP inhibitor, olaparib, results in enhanced anti-tumor activity in preclinical acute myeloid leukemia (AML) models. Blood. 2016;128(22):1645.
37.
Laszlo GS, Gudgeon CJ, Harrington KH, Dell'Aringa J, Newhall KJ, Means GD, et al. Cellular determinants for preclinical activity of a novel CD33/CD3 bispecific T-cell engager (BiTE) antibody, AMG 330, against human AML. Blood. 2014;123(4):554–61.CrossRefPubMedPubMedCentral
38.
Borthakur G, Rosenblum MG, Talpaz M, Daver N, Ravandi F, Faderl S, et al. Phase 1 study of an anti-CD33 immunotoxin, humanized monoclonal antibody M195 conjugated to recombinant gelonin (HUM-195/rGEL), in patients with advanced myeloid malignancies. Haematologica. 2013;98(2):217–21.CrossRefPubMedPubMedCentral
39.
Noordhuis P, Terwinjn M, Rutten AP, Smit L, Ossenkoppele GJ, Schuurhuis GJ. Targeting of CLEC12A in acute myeloid leukemia by antibody-drug-conjugates and bispecific CLL-1 × CD3 BiTE antibody. Blood. 2010;116:2890.
40.
Kim SY, Theunissen JW, Balibalos J, Liao-Chan S, Babcock MC, Wong T, et al. A novel antibody-drug conjugate targeting SAIL for the treatment of hematologic malignancies. Blood Cancer J. 2015;5, e316.CrossRefPubMedPubMedCentral
41.
Buckley SA, Walter RB. Antigen-specific immunotherapies for acute myeloid leukemia. Hematology Am Soc Hematol Educ Program. 2015;2015(1):584–95.PubMed
42.
Flynn MJ, van Berkel PH, Zammarchi F, Tyrer PC, Akarca AU, Janghra N, et al. Mechanistic and pharmacodynamic studies of adct-301, a pyrrolobenzodiazepine (PBD) dimer-containing antibody drug conjugate (ADC) targeting CD25-expressing hematological malignancies. Blood. 2015;126:1559.
43.
Rudra-Ganguly N, Lowe C, Virata C, Leavitt M, Jin L, Mendelsohn B, et al. AGS62P1, a novel anti-FLT3 antibody drug conjugate, employing site specific conjugation, demonstrates preclinical anti-tumor efficacy in AML tumor and patient derived xenografts. Blood. 2015;126:3806.
44.
Baeuerle PA, Reinhardt C. Bispecific T-cell engaging antibodies for cancer therapy. Cancer Res. 2009;69(12):4941–4.CrossRefPubMed
45.
Kontermann RE, Brinkmann U. Bispecific antibodies. Drug Discov Today. 2015;20(7):838–47.CrossRefPubMed
46.
Kantarjian H, Stein A, Gökbuget N, Fielding AK, Schuh AC, Ribera JM, et al. Blinatumomab versus chemotherapy for advanced acute lymphoblastic leukemia. N Engl J Med. 2017;376(9):836–47.CrossRefPubMed
47.
Aigner M, Feulner J, Schaffer S, Kischel R, Kufer P, Schneider K, et al. T lymphocytes can be effectively recruited for ex vivo and in vivo lysis of AML blasts by a novel CD33/CD3-bispecific BiTE antibody construct. Leukemia. 2013;27(5):1107–15.CrossRefPubMed
48.
Krupka C, Kufer P, Kischel R, Zugmaier G, Lichtenegger FS, Köhnke T, et al. Blockade of the PD-1/PD-L1 axis augments lysis of AML cells by the CD33/CD3-BiTE® antibody construct AMG 330: reversing a T-cell induced immune escape mechanism. Leukemia. 2015;30(2):484–91.CrossRefPubMed
49.
Arndt C, Feldmann A, von Bonin M, Cartellieri M, Ewen EM, Koristka S, et al. Costimulation improves the killing capability of T cells redirected to tumor cells expressing low levels of CD33: description of a novel modular targeting system. Leukemia. 2014;28(1):59–69.CrossRefPubMed
50.
Reusch U, Harrington KH, Gudgeon CJ, Fucek I, Ellwanger K, Weichel M, et al. Characterization of CD33/CD3 tetravalent bispecific tandem diabodies (TandAbs) for the treatment of acute myeloid leukemia. Clin Cancer Res. 2016;22(23):5829–38.CrossRefPubMed
51.
Moore PA, Zhang W, Rainey GJ, Burke S, Li H, Huang L, et al. Application of dual affinity retargeting molecules to achieve optimal redirected T-cell killing of B-cell lymphoma. Blood. 2011;117(17):4542–51.CrossRefPubMed
52.
Rader C. DARTs take aim at BiTEs. Blood. 2011;117(17):4403–4.CrossRefPubMed
53.
AL Hussaini MH, Ritchey J, Rettig MP, Eissenberg L, Uy GL, Chichili G, et al. Targeting CD123 in leukemic stem cells using dual affinity re-targeting molecules (DARTs®) [abstract]. Blood. 2013;122(21):360.
54.
Chichili GR, Huang L, Li H, Burke S, He L, Tang Q, et al. A CD3xCD123 bispecific DART for redirecting host T cells to myelogenous leukemia: preclinical activity and safety in nonhuman primates. Sci Transl Med. 2015;7(289):289ra82.CrossRefPubMed
55.
Chu SY, Pong E, Chen H, Phung S, Chan EW, Endo NA, et al. Immunotherapy with long-lived anti-CD123 × anti-CD3 bispecific antibodies stimulates potent T cell-mediated killing of human AML cell lines and of CD123+ cells in monkeys: a potential therapy for acute myelogenous leukemia. Blood. 2014;124:2316.CrossRef
56.
Forslund A, Syed K, Axel A, McDaid R, Li Y, Ballesteros J, et al. Ex vivo activity profile of the CD123xCD3 Duobody® antibody JNJ-63709178 against primary acute myeloid leukemia bone marrow samples. Blood. 2016;128:2875.CrossRef
57.
Gaudet F, Nemeth JF, McDaid R, Li Y, Harman B, Millar H, et al. Development of a CD123xCD3 bispecific antibody (JNJ-63709178) for the treatment of acute myeloid leukemia (AML). Blood. 2016;128:2824.CrossRef
58.
Bakker ABH, Oudenrijn SVD, Bakker AQ, Feller N, Meijer MV, Bia JA, et al. C-type lectin-like molecule-1: a novel myeloid cell surface marker associated with acute myeloid leukemia. Cancer Res. 2004;64(22):8443–50.CrossRefPubMed
59.
Rhenen AV, Dongen GAMSV, Kelder A, Rombouts EJ, Feller N, Moshaver B, et al. The novel AML stem cell–associated antigen CLL-1 aids in discrimination between normal and leukemic stem cells. Blood. 2007;110(7):2659–66.CrossRefPubMed
60.
Van Loo PF, 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.
61.
Roskopf CC, Braciak TA, Fenn NC, Kobold S, Fey GH, Hopfner KP, et al. Dual-targeting triplebody 33-3-19 mediates selective lysis of biphenotypic CD19+ CD33+ leukemia cells. Oncotarget. 2016;7(16):22579–89.CrossRefPubMedPubMedCentral
62.
Polu KR, Lowman HB. Probody therapeutics for targeting antibodies to diseased tissue. Expert Opin Biol Ther. 2014;14(8):1049–53.CrossRefPubMed
63.
LaPorte SL, Hostetter DR, Wong L, Razo J, Diep L, White CW, et al. CD3-EGFR bispecific Probody™ therapeutics induced tumor regressions and increased therapeutic window in preclinical studies. Proceedings of the AACR-NCI-EORTC International Conference: Molecular Targets and Cancer Therapeutics. 2015;14:Abstract nr A203.
64.
Zugmaier G, Klinger M, Schmidt M, Subklewe M. Clinical overview of anti-CD19 BiTE(®) and ex vivo data from anti-CD33 BiTE(®) as examples for retargeting T cells in hematologic malignancies. Mol Immunol. 2015.
65.
Köhnke T, Krupka C, Tischer J, Knösel T, Subklewe M. Increase of PD-L1 expressing B-precursor ALL cells in a patient resistant to the CD19/CD3-bispecific T cell engager antibody blinatumomab. J Hematol Oncol. 2015;8:111.CrossRefPubMedPubMedCentral
66.
Gross G, Waks T, Eshhar Z. Expression of immunoglobulin-T-cell receptor chimeric molecules as functional receptors with antibody-type specificity. Proc Natl Acad Sci U S A. 1989;86(24):10024–8.CrossRefPubMedPubMedCentral
67.
Finney HM, Lawson AD, Bebbington CR, Weir AN. Chimeric receptors providing both primary and costimulatory signaling in T cells from a single gene product. J Immunol. 1998;161(6):2791–7.PubMed
68.
Turtle CJ, Hanafi LA, Berger C, Gooley TA, Cherian S, Hudecek M, et al. CD19 CAR-T cells of defined CD4+:CD8+ composition in adult B cell ALL patients. J Clin Invest. 2016;126(6):2123–38.CrossRefPubMedPubMedCentral
69.
Locke FL, Neelapu SS, Bartlett NL, Siddiqi T, Chavez JC, Hosing CM, et al. Phase 1 results of ZUMA-1: a multicenter study of KTE-C19 anti-CD19 CAR T cell therapy in refractory aggressive lymphoma. Mol Ther. 2017;25(1):285–95.CrossRefPubMed
70.
O’Leary M. FDA pilot project to develop a clinical database to examine safety in trials using CAR T-cells. http://​www.​ema.​europa.​eu/​docs/​en_​GB/​document_​library/​Presentation/​2016/​12/​WC500217505.​pdf. Accessed 6 Mar 2017.
71.
Juno Therapeutics, Inc. Juno Therapeutics reports fourth quarter and 2016 financial results. http://​ir.​junotherapeutics​.​com/​phoenix.​zhtml?​c=​253828&​p=​irol-newsArticle_​Print&​ID=​2250772. Accessed 6 Mar 2017.
72.
Porter DL, Hwang WT, Frey NV, Lacey SF, Shaw PA, Loren AW, et al. Chimeric antigen receptor T cells persist and induce sustained remissions in relapsed refractory chronic lymphocytic leukemia. Sci Transl Med. 2015;7(303):303ra139.CrossRefPubMed
73.
Chevallier P, Robillard N, Ayari S, Guillaume T, Delaunay J, Mechinaud F, et al. Persistence of CD33 expression at relapse in CD33(+) acute myeloid leukaemia patients after receiving gemtuzumab in the course of the disease. Br J Haematol. 2008;143(5):744–6.CrossRefPubMed
74.
O'Hear C, Heiber JF, Schubert I, Fey G, Geiger TL. Anti-CD33 chimeric antigen receptor targeting of acute myeloid leukemia. Haematologica. 2015;100(3):336–44.CrossRefPubMedPubMedCentral
75.
Kenderian SS, Ruella M, Shestova O, Klichinsky M, Aikawa V, Morrissette JJ, et al. CD33-specific chimeric antigen receptor T cells exhibit potent preclinical activity against human acute myeloid leukemia. Leukemia. 2015;29(8):1637–47.CrossRefPubMedPubMedCentral
76.
Mardiros A, Dos Santos C, McDonald T, Brown CE, Wang X, Budde LE, et al. T cells expressing CD123-specific chimeric antigen receptors exhibit specific cytolytic effector functions and anti-tumor effects against human acute myeloid leukemia. Blood. 2013;122(18):3138–48.CrossRefPubMedPubMedCentral
77.
Tettamanti S, Marin V, Pizzitola I, Magnani CF, Giordano Attianese GM, Cribioli E, et al. Targeting of acute myeloid leukaemia by cytokine-induced killer cells redirected with a novel CD123-specific chimeric antigen receptor. Br J Haematol. 2013;161(3):389–401.CrossRefPubMed
78.
Pizzitola I, Anjos-Afonso F, Rouault-Pierre K, Lassailly F, Tettamanti S, Spinelli O, et al. Chimeric antigen receptors against CD33/CD123 antigens efficiently target primary acute myeloid leukemia cells in vivo. Leukemia. 2014;28(8):1596–605.CrossRefPubMed
79.
Thokala R, Olivares S, Mi T, Maiti S, Deniger D, Huls H, et al. Redirecting specificity of T cells using the sleeping beauty system to express chimeric antigen receptors by mix-and-matching of VL and VH domains targeting CD123+ tumors. PLoS One. 2016;11(8), e0159477.CrossRefPubMedPubMedCentral
80.
Gill S, Tasian SK, Ruella M, Shestova O, Li Y, Porter DL, et al. Preclinical targeting of human acute myeloid leukemia and myeloablation using chimeric antigen receptor-modified T cells. Blood. 2014;123(15):2343–54.CrossRefPubMedPubMedCentral
81.
Casucci M, di Nicolis Robilant B, Falcone L, Camisa B, Norelli M, Genovese P, et al. CD44v6-targeted T cells mediate potent antitumor effects against acute myeloid leukemia and multiple myeloma. Blood. 2013;122(20):3461.CrossRefPubMed
82.
Kenderian SS, Ruella M, Shestova O, Klichinsky M, Kim MY, Soderquist C, 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.
83.
Chien CD, Sauter CT, Ishii K, Nguyen SM, Shen F, Tasian SK, et al. Preclinical development of FLT3-redirected chimeric antigen receptor T cell immunotherapy for acute myeloid leukemia. Blood. 2016;128:1072.CrossRef
84.
Lynn RC, Feng Y, Schutsky K, Poussin M, Kalota A, Dimitrov DS, et al. High-affinity FRβ-specific CAR T cells eradicate AML and normal myeloid lineage without HSC toxicity. Leukemia. 2016;30(6):1355–64.CrossRefPubMedPubMedCentral
85.
Peinert S, Prince HM, Guru PM, Kershaw MH, Smyth MJ, Trapani JA, et al. Gene-modified T cells as immunotherapy for multiple myeloma and acute myeloid leukemia expressing the Lewis Y antigen. Gene Ther. 2010;17(5):678–86.CrossRefPubMed
86.
Chang YH, Connolly J, Shimasaki N, Mimura K, Kono K, Campana D. A chimeric receptor with NKG2D specificity enhances natural killer cell activation and killing of tumor cells. Cancer Res. 2013;73(6):1777–86.CrossRefPubMed
87.
Ma Q, Garber HR, Lu S, He H, Tallis E, Ding X, et al. A novel TCR-like CAR with specificity for PR1/HLA-A2 effectively targets myeloid leukemia in vitro when expressed in human adult peripheral blood and cord blood T cells. Cytotherapy. 2016;18(8):985–94.CrossRefPubMed
88.
Ritchie DS, Neeson PJ, Khot A, Peinert S, Tai T, Tainton K, et al. Persistence and efficacy of second generation CAR T cell against the LeY antigen in acute myeloid leukemia. Mol Ther. 2013;21(11):2122–9.CrossRefPubMedPubMedCentral
89.
Wang QS, Wang Y, Lv HY, Han QW, Fan H, Guo B, et al. Treatment of CD33-directed chimeric antigen receptor-modified T cells in one patient with relapsed and refractory acute myeloid leukemia. Mol Ther. 2014;23(1):184–91.CrossRefPubMedPubMedCentral
90.
Guzman ML, Sugita M, Zong H, Ewing-Crystal N, Trujillo-Alonso V, Mencia-Trinchant N, et al. Allogeneic Tcrα/β deficient CAR T-cells targeting CD123 prolong overall survival of AML patient-derived xenografts. Blood. 2016;128(22):765.
91.
Lanitis E, Poussin M, Klattenhoff AW, Song D, Sandaltzopoulos R, June CH, et al. Chimeric antigen receptor T cells with dissociated signaling domains exhibit focused antitumor activity with reduced potential for toxicity in vivo. Cancer Immunol Res. 2013;1(1):43–53.CrossRefPubMedPubMedCentral
92.
Zah E, Lin MY, Silva-Benedict A, Jensen MC, Chen YY. T cells expressing CD19/CD20 bispecific chimeric antigen receptors prevent antigen escape by malignant B cells. Cancer Immunol Res. 2016;4(6):498–508.CrossRefPubMedPubMedCentral
93.
Arndt C, Feldmann A, Koristka S, Cartellieri M, von Bonin M, Ehninger A, et al. Improved killing of AML blasts by dual-targeting of CD123 and CD33 via UniTARG a novel antibody-based modular T cell retargeting system. Blood. 2015;126:2565.
94.
Arcangeli S, Bardelli M, Tettamanti S, Rotiroti MC, Simonelli L, Magnani CF, et al. Unraveling the efficacy and safety profiles of anti-CD123 chimeric antigen receptors (CARs) in a model of acute myeloid leukemia immunotherapy by investigating CAR binding affinity and density variables. Blood. 2015;126:1359.
95.
Resetca D, Neschadim A, Medin JA. Engineering hematopoietic cells for cancer immunotherapy: strategies to address safety and toxicity concerns. J Immunother. 2016;39(7):249–59.CrossRefPubMed
96.
Alatrash G, Daver N, Mittendorf EA. Targeting immune checkpoints in hematologic malignancies. Pharmacol Rev. 2016;68(4):1014–25.CrossRefPubMed
97.
Berger R, Rotem-Yehudar R, Slama G, Landes S, Kneller A, Leiba M, et al. 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(10):3044–51.CrossRefPubMed
98.
Yang H, Bueso-Ramos C, DiNardo C, Estecio MR, Davanlou M, Geng QR, 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(6):1280–8.CrossRefPubMed
99.
Daver N, Basu S, Garcia-Manero G, Cortes JE, Ravandi F, Jabbour EJ, 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(22):763.
100.
Sabado RL, Balan S, Bhardwaj N. Dendritic cell-based immunotherapy. Cell Res. 2017;27(1):74–95.CrossRefPubMed
101.
Rosenblatt J, Stone RM, Uhl L, Neuberg D, Joyce R, Levine JD, et al. Individualized vaccination of AML patients in remission is associated with induction of antileukemia immunity and prolonged remissions. Sci Transl Med. 2016;8(368):368ra171.CrossRefPubMed
102.
Van Tendeloo VF, Van de Velde A, Van Driessche A, Cools N, Anguille S, Ladell K, et al. Induction of complete and molecular remissions in acute myeloid leukemia by Wilms’ tumor 1 antigen-targeted dendritic cell vaccination. Proc Natl Acad Sci U S A. 2010;107(31):13824–9.CrossRefPubMedPubMedCentral
103.
Spranger S, Javorovic M, Bürdek M, Wilde S, Mosetter B, Tippmer S, et al. Generation of Th1-polarizing dendritic cells using the TLR7/8 agonist CL075. J Immunol. 2010;185(1):738–47.CrossRefPubMed
104.
Beck B, Dörfel D, Lichtenegger FS, Geiger C, Lindner L, Merk M, et al. Effects of TLR agonists on maturation and function of 3-day dendritic cells from AML patients in complete remission. J Transl Med. 2011;9:151.CrossRefPubMedPubMedCentral
105.
Lichtenegger FS, Mueller K, Otte B, Beck B, Hiddemann W, Schendel DJ, et al. CD86 and IL-12p70 are key players for T helper 1 polarization and natural killer cell activation by toll-like receptor-induced dendritic cells. PLoS One. 2012;7(9), e44266.CrossRefPubMedPubMedCentral
106.
Subklewe M, Geiger C, Lichtenegger FS, Javorovic M, Kvalheim G, Schendel DJ, et al. New generation dendritic cell vaccine for immunotherapy of acute myeloid leukemia. Cancer Immunol Immunother. 2014;63(10):1093–103.CrossRefPubMed
107.
Lichtenegger FS, Deiser K, Rothe M, Schnorfeil FM, Krupka C, Augsberger C, et al. Induction of antigen-specific T-cell responses through dendritic cell vaccination in AML: results of a phase I/II trial and ex vivo enhancement by checkpoint blockade. Blood. 2016;128(22):764.
108.
Van den Bergh J, Willemen Y, Lion E, Van Acker H, De Reu H, Anguille S, et al. Transpresentation of interleukin-15 by IL-15/IL-15Rα mRNA-engineered human dendritic cells boosts antitumoral natural killer cell activity. Oncotarget. 2015;6(42):44123–33.CrossRefPubMedPubMedCentral
109.
Willemen Y, Van den Bergh JM, Bonte SM, Anguille S, Heirman C, Stein BM, et al. The tumor-associated antigen RHAMM (HMMR/CD168) is expressed by monocyte-derived dendritic cells and presented to T cells. Oncotarget. 2016;7(45):73960–70.PubMedPubMedCentral
110.
Perna F, Berman S, Mansilla-Soto J, Hamieh M, Juthani R, Soni R, et al. Probing the AML surfaceome for chimeric antigen receptor (CAR) targets. Blood. 2016;128(22):526.