Skip to main content

Thank you for visiting nature.com. You are using a browser version with limited support for CSS. To obtain the best experience, we recommend you use a more up to date browser (or turn off compatibility mode in Internet Explorer). In the meantime, to ensure continued support, we are displaying the site without styles and JavaScript.

  • Original Article
  • Published:

Targeting of acute myeloid leukemia in vitro and in vivo with an anti-CD123 mAb engineered for optimal ADCC

Abstract

Acute myeloid leukemia (AML) is a biologically heterogeneous group of related diseases in urgent need of better therapeutic options. Despite this heterogeneity, overexpression of the interleukin (IL)-3 receptor α-chain (IL-3 Rα/CD123) on both the blast and leukemic stem cell (LSC) populations is a common occurrence, a finding that has generated wide interest in devising new therapeutic approaches that target CD123 in AML patients. We report here the development of CSL362, a monoclonal antibody to CD123 that has been humanized, affinity-matured and Fc-engineered for increased affinity for human CD16 (FcγRIIIa). In vitro studies demonstrated that CSL362 potently induces antibody-dependent cell-mediated cytotoxicity of both AML blasts and CD34+CD38CD123+ LSC by NK cells. Importantly, CSL362 was highly effective in vivo reducing leukemic cell growth in AML xenograft mouse models and potently depleting plasmacytoid dendritic cells and basophils in cynomolgus monkeys. Significantly, we demonstrated CSL362-dependent autologous depletion of AML blasts ex vivo, indicating that CSL362 enables the efficient killing of AML cells by the patient’s own NK cells. These studies offer a new therapeutic option for AML patients with adequate NK-cell function and warrant the clinical development of CSL362 for the treatment of AML.

This is a preview of subscription content, access via your institution

Access options

Buy this article

Prices may be subject to local taxes which are calculated during checkout

Figure 1
Figure 2
Figure 3
Figure 4
Figure 5

Similar content being viewed by others

References

  1. Roboz GJ . Current treatment of acute myeloid leukemia. Curr Opin Oncol 2012; 24: 711–719.

    Article  CAS  Google Scholar 

  2. Bonnet D, Dick JE . Human acute myeloid leukemia is organized as a hierarchy that originates from a primitive hematopoietic cell. Nat Med 1997; 3: 730–737.

    Article  CAS  Google Scholar 

  3. Lapidot T, Sirard C, Vormoor J, Murdoch B, Hoang T, Caceres-Cortes J et al. A cell initiating human acute myeloid leukaemia after transplantation into SCID mice. Nature 1994; 367: 645–648.

    Article  CAS  Google Scholar 

  4. Hope KJ, Jin L, Dick JE . Acute myeloid leukemia originates from a hierarchy of leukemic stem cell classes that differ in self-renewal capacity. Nat Immunol 2004; 5: 738–743.

    Article  CAS  Google Scholar 

  5. Guzman ML, Neering SJ, Upchurch D, Grimes B, Howard DS, Rizzieri DA et al. Nuclear factor-kappaB is constitutively activated in primitive human acute myelogenous leukemia cells. Blood 2001; 98: 2301–2307.

    Article  CAS  Google Scholar 

  6. Guan Y, Gerhard B, Hogge DE . Detection, isolation, and stimulation of quiescent primitive leukemic progenitor cells from patients with acute myeloid leukemia (AML). Blood 2003; 101: 3142–3149.

    Article  CAS  Google Scholar 

  7. Ishikawa F, Yoshida S, Saito Y, Hijikata A, Kitamura H, Tanaka S et al. Chemotherapy-resistant human AML stem cells home to and engraft within the bone-marrow endosteal region. Nat Biotechnol 2007; 25: 1315–1321.

    Article  CAS  Google Scholar 

  8. Testa U, Riccioni R, Militi S, Coccia E, Stellacci E, Samoggia P et al. Elevated expression of IL-3Ralpha in acute myelogenous leukemia is associated with enhanced blast proliferation, increased cellularity, and poor prognosis. Blood 2002; 100: 2980–2988.

    Article  CAS  Google Scholar 

  9. Muñoz L, Nomdedéu JF, López O, Carnicer MJ, Bellido M, Aventín A et al. Interleukin-3 receptor alpha chain (CD123) is widely expressed in hematologic malignancies. Haematologica 2001; 86: 1261–1269.

    Google Scholar 

  10. De Waele M, Renmans W, Vander Gucht K, Jochmans K, Schots R, Otten J et al. Growth factor receptor profile of CD34+ cells in AML and B-lineage ALL and in their normal bone marrow counterparts. Eur J Haematol 2001; 66: 178–187.

    Article  CAS  Google Scholar 

  11. Djokic M, Bjorklund E, Blennow E, Mazur J, Soderhall S, Porwit A . Overexpression of CD123 correlates with the hyperdiploid genotype in acute lymphoblastic leukemia. Haematologica 2009; 94: 1016–1019.

    Article  CAS  Google Scholar 

  12. Hassanein NM, Alcancia F, Perkinson KR, Buckley PJ, Lagoo AS . Distinct expression patterns of CD123 and CD34 on normal bone marrow B-cell precursors (“hematogones”) and B lymphoblastic leukemia blasts. Am J Clin Pathol 2009; 132: 573–580.

    Article  Google Scholar 

  13. Jordan CT, Upchurch D, Szilvassy SJ, Guzman ML, Howard DS, Pettigrew AL et al. The interleukin-3 receptor alpha chain is a unique marker for human acute myelogenous leukemia stem cells. Leukemia 2000; 14: 1777–1784.

    Article  CAS  Google Scholar 

  14. Sun Q, Woodcock JM, Rapoport A, Stomski FC, Korpelainen EI, Bagley CJ et al. Monoclonal antibody 7G3 recognizes the N-terminal domain of the human interleukin-3 (IL-3) receptor alpha-chain and functions as a specific IL-3 receptor antagonist. Blood 1996; 87: 83–92.

    CAS  Google Scholar 

  15. Jin L, Lee EM, Ramshaw HS, Busfield SJ, Peoppl AG, Wilkinson L et al. Monoclonal antibody-mediated targeting of CD123, IL-3 receptor & alpha; chain. Cell Stem Cell 2009; 5: 31–42.

    Article  CAS  Google Scholar 

  16. Roberts AW, He S, Ritchie D, Hertzberg MS, Kerridge I, Durrant ST et al. A phase I study of anti-CD123 monoclonal antibody (mAb) CSL360 targeting leukemia stem cells (LSC) in AML. J Clin Oncol 2010; 28: e13012.

    Article  Google Scholar 

  17. Cartron G, Dacheux L, Salles G, Solal-Celigny P, Bardos P, Colombat P et al. Therapeutic activity of humanized anti-CD20 monoclonal antibody and polymorphism in IgG Fc receptor FcgammaRIIIa gene. Blood 2002; 99: 754–758.

    Article  CAS  Google Scholar 

  18. Weng W-K, Levy R . Two immunoglobulin G fragment C receptor polymorphisms independently predict response to rituximab in patients with follicular lymphoma. J Clin Oncol 2003; 21: 3940–3947.

    Article  CAS  Google Scholar 

  19. Dall'Ozzo S, Tartas S, Paintaud G, Cartron G, Colombat P, Bardos P et al. Rituximab-dependent cytotoxicity by natural killer cells: influence of FCGR3A polymorphism on the concentration-effect relationship. Cancer Res 2004; 64: 4664–4669.

    Article  CAS  Google Scholar 

  20. Lazar GA, Dang W, Karki S, Vafa O, Peng JS, Hyun L et al. Engineered antibody Fc variants with enhanced effector function. Proc Natl Acad Sci USA 2006; 103: 4005–4010.

    Article  CAS  Google Scholar 

  21. Shields RL, Lai J, Keck R, O'Connell LY, Hong K, Meng YG et al. Lack of fucose on human IgG1 N-linked oligosaccharide improves binding to human Fc gamma RIII and antibody-dependent cellular toxicity. J Biol Chem 2002; 277: 26733–26740.

    Article  CAS  Google Scholar 

  22. Horton HM, Bernett MJ, Peipp M, Pong E, Karki S, Chu SY et al. Fc-engineered anti-CD40 antibody enhances multiple effector functions and exhibits potent in vitro and in vivo antitumor activity against hematologic malignancies. Blood 2010; 116: 3004–3012.

    Article  CAS  Google Scholar 

  23. Ito A, Ishida T, Utsunomiya A, Sato F, Mori F, Yano H et al. Defucosylated anti-CCR4 monoclonal antibody exerts potent ADCC against primary ATLL cells mediated by autologous human immune cells in NOD/Shi-scid, IL-2R null mice in vivo. J Immunol 2009; 183: 4782–4791.

    Article  CAS  Google Scholar 

  24. McEarchern JA, Oflazoglu E, Francisco L, McDonagh CF, Gordon KA, Stone I et al. Engineered anti-CD70 antibody with multiple effector functions exhibits in vitro and in vivo antitumor activities. Blood 2006; 109: 1185–1192.

    Article  Google Scholar 

  25. Cardarelli PM, Rao-Naik C, Chen S, Huang H, Pham A, Moldovan-Loomis M-C et al. A nonfucosylated human antibody to CD19 with potent B-cell depletive activity for therapy of B-cell malignancies. Cancer Immunol Immunother 2009; 59: 257–265.

    Article  Google Scholar 

  26. Bowles JA, Weiner GJ . CD16 polymorphisms and NK activation induced by monoclonal antibody-coated target cells. J Immunol Methods 2005; 304: 88–99.

    Article  CAS  Google Scholar 

  27. Koene HR, Kleijer M, Algra J, Roos D, Borne von dem AE, de Haas M . Fc gammaRIIIa-158V/F polymorphism influences the binding of IgG by natural killer cell Fc gammaRIIIa, independently of the Fc gammaRIIIa-48L/R/H phenotype. Blood 1997; 90: 1109–1114.

    CAS  Google Scholar 

  28. Penack O, Gentilini C, Fischer L, Asemissen AM, Scheibenbogen C, Thiel E et al. CD56dimCD16neg cells are responsible for natural cytotoxicity against tumor targets. Leukemia 2005; 19: 835–840.

    Article  CAS  Google Scholar 

  29. Tan P, Mitchell DA, Buss TN, Holmes MA, Anasetti C, Foote J . “Superhumanized” antibodies: reduction of immunogenic potential by complementarity-determining region grafting with human germline sequences: application to an anti-CD28. J Immunol 2002; 169: 1119–1125.

    Article  CAS  Google Scholar 

  30. Kopsidas G, Roberts AS, Coia G, Streltsov VA, Nuttall SD . In vitro improvement of a shark IgNAR antibody by Qβ replicase mutation and ribosome display mimics in vivo affinity maturation. Immunol Lett 2006; 107: 163–168.

    Article  CAS  Google Scholar 

  31. Zalevsky J, Leung IWL, Karki S, Chu SY, Zhukovsky EA, Desjarlais JR et al. The impact of Fc engineering on an anti-CD19 antibody: increased Fc receptor affinity enhances B-cell clearing in nonhuman primates. Blood 2009; 113: 3735–3743.

    Article  CAS  Google Scholar 

  32. Warren HS . Target-induced natural killer cell loss as a measure of NK cell responses. J Immunol Methods 2011; 370: 86–92.

    Article  CAS  Google Scholar 

  33. Meropol NJ, Barresi GM, Fehniger TA, Hitt J, Franklin M, Caligiuri MA . Evaluation of natural killer cell expansion and activation in vivo with daily subcutaneous low-dose interleukin-2 plus periodic intermediate-dose pulsing. Cancer Immunol Immunother 1998; 46: 318–326.

    Article  CAS  Google Scholar 

  34. Stone RM . Is it time to revisit standard post-remission therapy? Best Pract Res Clin Haematol 2012; 25: 437–441.

    Article  Google Scholar 

  35. Horton SJ, Huntly BJP . Recent advances in acute myeloid leukemia stem cell biology. Haematologica 2012; 97: 966–974.

    Article  CAS  Google Scholar 

  36. Yalcintepe L, Frankel AE, Hogge DE . Expression of interleukin-3 receptor subunits on defined subpopulations of acute myeloid leukemia blasts predicts the cytotoxicity of diphtheria toxin interleukin-3 fusion protein against malignant progenitors that engraft in immunodeficient mice. Blood 2006; 108: 3530–3537.

    Article  CAS  Google Scholar 

  37. Frankel A, Liu J-S, Rizzieri D, Hogge D . Phase I clinical study of diphtheria toxin-interleukin 3 fusion protein in patients with acute myeloid leukemia and myelodysplasia. Leuk Lymphoma 2008; 49: 543–553.

    Article  CAS  Google Scholar 

  38. Konopleva M, Hogge DE, Rizzieri DA, Cirrito TP, Kornblau SM, Borthakur G et al. SL-401, a targeted therapy directed to the interleukin-3 receptor present on leukemia blasts and cancer stem cells, is active as a single agent in patients with advanced AML. Blood 2012; 120, abstract 3625.

    Article  Google Scholar 

  39. Tettamanti S, Marin V, Pizzitola I, Magnani CF, Giordano Attianese GMP, 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: 389–401.

    Article  CAS  Google Scholar 

  40. Kuo SR, Wong L, Liu JS . Engineering a CD123xCD3 bispecific scFv immunofusion for the treatment of leukemia and elimination of leukemia stem cells. Protein Eng Des Sel 2012; 25: 561–570.

    Article  CAS  Google Scholar 

  41. Kugler M, Stein C, Kellner C, Mentz K, Saul D, Schwenkert M 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.

    Article  Google Scholar 

  42. Kurai J, Chikumi H, Hashimoto K, Yamaguchi K, Yamasaki A, Sako T et al. Antibody-dependent cellular cytotoxicity mediated by Cetuximab against lung cancer cell lines. Clin Cancer Res 2007; 13: 1552–1561.

    Article  CAS  Google Scholar 

  43. Dauguet N, Récher C, Demur C, Fournie J-J, Poupot M, Poupot R . Pre-eminence and persistence of immature natural killer cells in acute myeloid leukemia patients in first complete remission. Am J Hematol 2011; 86: 209–213.

    Article  Google Scholar 

  44. Barrett J, Le Blanc K . Cancer chemotherapy and immune regulation. Am J Immunol 2009; 5: 8–16.

    Article  CAS  Google Scholar 

  45. Rey J, Anfossi N, Andre P, Boher J-M, Orlanducci F, Breso V et al. Natural killer cells recovery after consolidation chemotherapy in elderly patients with acute myeloid leukemia (AML). Blood 2011; 118, abstract 2189.

    Google Scholar 

  46. Lion E, Willemen Y, Berneman ZN, Van Tendeloo VFI, Smits ELJ . Natural killer cell immune escape in acute myeloid leukemia. Leukemia 2012; 26: 2019–2026.

    Article  CAS  Google Scholar 

  47. Bryceson YT, March ME, Ljunggren H-G, Long EO . Activation, coactivation, and costimulation of resting human natural killer cells. Immunol Rev 2006; 214: 73–91.

    Article  CAS  Google Scholar 

  48. Costello RT, Sivori S, Marcenaro E, Lafage-Pochitaloff M, Mozziconacci M-J, Reviron D et al. Defective expression and function of natural killer cell-triggering receptors in patients with acute myeloid leukemia. Blood 2002; 99: 3661–3667.

    Article  CAS  Google Scholar 

Download references

Acknowledgements

We would like to the following for their contributions: Bill Panagopoulos and Melanie Pudney (IMVS), Laura McMillan, Kirstee Martin, Daria Kurtov, Matt Hardy and members of the CSL Research Department for excellent technical assistance. Naomi Sprigg for patient sample collection. Hayley Ramshaw is supported by the Peter Nelson Leukaemia Research Fund. Children’s Cancer Institute Australia for Medical Research is affiliated with the University of New South Wales and the Sydney Children’s Hospitals Network. Andrew Roberts is supported by funding from NHMRC (637309, 1016647) and the Leukemia and Lymphoma Society.

Author information

Authors and Affiliations

Authors

Corresponding author

Correspondence to S J Busfield.

Ethics declarations

Competing interests

CSL Limited is developing the CSL362 monoclonal antibody. AFL, HSR, EML and RBL received research funding from CSL Limited. RBL was and AFL is a consultant for CSL Limited. SJB, MB, MW, SG, HB, CP, LF, GV and ADN are employed by CSL Limited. AWR, SZH and DT declare no conflict of interest.

Additional information

Author contributions

MB, MW, HSR, HB, EML and SG designed and performed the experiments and analyzed the data. SJB designed the research, reviewed the data and wrote the manuscript. CP and LF contributed vital new reagents. GV, RBL, AFL, AWR and ADN were significantly involved in experimental design, data interpretation and manuscript preparation. SZH and DT contributed clinical samples and critically reviewed the manuscript.

Supplementary Information accompanies this paper on the Leukemia website

Supplementary information

Rights and permissions

Reprints and permissions

About this article

Cite this article

Busfield, S., Biondo, M., Wong, M. et al. Targeting of acute myeloid leukemia in vitro and in vivo with an anti-CD123 mAb engineered for optimal ADCC. Leukemia 28, 2213–2221 (2014). https://doi.org/10.1038/leu.2014.128

Download citation

  • Received:

  • Revised:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/leu.2014.128

This article is cited by

Search

Quick links