Post-hoc Analysis of Pharmacodynamics and Single-Agent Activity of CD3xCD123 Bispecific Antibody APVO436 in Relapsed/Refractory AML and MDS Resistant to HMA or Venetoclax Plus HMA

APVO436 is a recombinant bispecific antibody designed to direct host cytotoxic T-cells to CD123-expressing blast cells in patients with hematologic malignancies. APVO436 showed promising tolerability and single-agent activity in relapsed or refractory (R/R) acute myeloid leukemia (AML) and myelodysplastic syndrome (MDS). The primary purpose of this post-hoc analysis was to evaluate the therapeutic and pharmacodynamic effects of APVO436 in 14 R/R AML/MDS patients who had failed treatment with hypomethylating agents (HMA) or venetoclax plus HMA prior to being enrolled in the APVO436 Phase 1 dose-escalation study that was recently completed. Eight of these 14 patients had R/R AML and had failed treatment with HMA (N=2) or venetoclax plus HMA (N=6). The remaining 6 patients had R/R MDS and had also failed treatment with HMA (N=5) or venetoclax plus HMA (N=1). They were treated with APVO436 at submicrogram dose levels >0.08 mcg/kg that were active in preclinical NOD/SCID mouse xenograft models of AML. APVO436 activated patients’ T-cells as evidenced by reduced numbers of circulating CD123+CD34+ and CD33+CD34+ peripheral blasts. Single-agent activity was observed at dose levels ranging from 0.1 mcg/kg to 0.7 mcg/kg in 4 R/R AML patients (50%), including 3 patients with prolonged stable disease (SD) and one patient with complete remission (CR). Likewise, 3 MDS patients had SD (50%) and 3 additional MDS patients (50%) had a marrow CR at dose levels ranging from 0.1 mcg/kg to 0.8 mcg/kg. The median survival for the combined group of 14 R/R AML/MDS patients was 282 days. This early evidence of single-agent activity of APVO436 in R/R AML/MDS patients who failed HMA with or without venetoclax provides proof of concept supporting its in vivo immunomodulatory and anti-leukemic activity and warrants further investigation of its clinical impact potential.

[1]  F. Uckun,et al.  Risk, Characteristics and Biomarkers of Cytokine Release Syndrome in Patients with Relapsed/Refractory AML or MDS Treated with CD3xCD123 Bispecific Antibody APVO436 , 2021, Cancers.

[2]  F. Uckun,et al.  A Clinical Phase 1B Study of the CD3xCD123 Bispecific Antibody APVO436 in Patients with Relapsed/Refractory Acute Myeloid Leukemia or Myelodysplastic Syndrome , 2021, Cancers.

[3]  Tamer Othman,et al.  Venetoclax-based combinations for the treatment of newly diagnosed acute myeloid leukemia. , 2021, Future Oncology.

[4]  M. Konopleva,et al.  Immunotherapy in Acute Myeloid Leukemia: Where We Stand , 2021, Frontiers in Oncology.

[5]  N. Daver,et al.  T-cell-based immunotherapy of acute myeloid leukemia: current concepts and future developments , 2021, Leukemia.

[6]  M. Minden,et al.  Venetoclax enhances T cell–mediated antileukemic activity by increasing ROS production , 2021, Blood.

[7]  M. Minden,et al.  Venetoclax enhances T cell-mediated anti-leukemic activity by increasing ROS production. , 2021, Blood.

[8]  J. Gabrilove,et al.  Safety and Efficacy: Clinical Experience of Venetoclax in Combination With Hypomethylating Agents in Both Newly Diagnosed and Relapsed/Refractory Advanced Myeloid Malignancies , 2021, HemaSphere.

[9]  A. Almasan,et al.  Targeting BCL-2 in B-cell malignancies and overcoming therapeutic resistance , 2020, Cell Death & Disease.

[10]  P. Vyas,et al.  New directions for emerging therapies in acute myeloid leukemia: the next chapter , 2020, Blood Cancer Journal.

[11]  S. Rutella,et al.  Flotetuzumab as Salvage Immunotherapy for Refractory Acute Myeloid Leukemia. , 2020, Blood.

[12]  A. Letai,et al.  Azacitidine and Venetoclax in Previously Untreated Acute Myeloid Leukemia. , 2020, The New England journal of medicine.

[13]  W. Blum,et al.  Treating acute myeloid leukemia in the modern era: A primer , 2020, Cancer.

[14]  David Lara-Astiaso,et al.  Immunogenomic identification and characterization of granulocytic myeloid derived suppressor cells in multiple myeloma. , 2020, Blood.

[15]  M. Konopleva,et al.  Advances in the Treatment of Acute Myeloid Leukemia: New Drugs and New Challenges. , 2020, Cancer discovery.

[16]  F. Uckun,et al.  A Phase 1B Clinical Study of Combretastatin A1 Diphosphate (OXi4503) and Cytarabine (ARA-C) in Combination (OXA) for Patients with Relapsed or Refractory Acute Myeloid Leukemia , 2019, Cancers.

[17]  C. Dinardo,et al.  How I treat acute myeloid leukemia in the era of new drugs. , 2019, Blood.

[18]  D. Jablonski,et al.  Abstract LB-199: APVO436, a bispecific anti-CD123 x anti-CD3 ADAPTIR™ molecule for redirected T-cell cytotoxicity with limited cytokine release, is well tolerated in repeat dose toxicology studies in cynomolgus macaques , 2019, Immunology.

[19]  W. Blum,et al.  Progress in the problem of relapsed or refractory acute myeloid leukemia , 2019, Current opinion in hematology.

[20]  F. Ferrara,et al.  Current Therapeutic Results and Treatment Options for Older Patients with Relapsed Acute Myeloid Leukemia , 2019, Cancers.

[21]  A. Letai,et al.  Venetoclax combined with decitabine or azacitidine in treatment-naive, elderly patients with acute myeloid leukemia. , 2019, Blood.

[22]  Austin E. Gillen,et al.  Venetoclax with azacitidine disrupts energy metabolism and targets leukemia stem cells in patients with acute myeloid leukemia , 2018, Nature Medicine.

[23]  A. D’Alessandro,et al.  Inhibition of Amino Acid Metabolism Selectively Targets Human Leukemia Stem Cells. , 2018, Cancer cell.

[24]  M. Konopleva,et al.  Clinical experience with the BCL2‐inhibitor venetoclax in combination therapy for relapsed and refractory acute myeloid leukemia and related myeloid malignancies , 2018, American journal of hematology.

[25]  A. Letai,et al.  Safety and preliminary efficacy of venetoclax with decitabine or azacitidine in elderly patients with previously untreated acute myeloid leukaemia: a non-randomised, open-label, phase 1b study. , 2018, The Lancet. Oncology.

[26]  R. Schlenk,et al.  Relapsed/refractory acute myeloid leukemia: any progress? , 2017, Current opinion in oncology.

[27]  A. Al-Mawali,et al.  CD34+CD38-CD123+ Cells Are Present in Virtually All Acute Myeloid Leukaemia Blasts: A Promising Single Unique Phenotype for Minimal Residual Disease Detection , 2017, Acta Haematologica.

[28]  Bob Löwenberg,et al.  Diagnosis and management of AML in adults: 2017 ELN recommendations from an international expert panel. , 2017, Blood.

[29]  James M Wilson,et al.  The Next Chapter. , 2015, Human gene therapy.

[30]  J. Dipersio,et al.  Targeting CD123 In Leukemic Stem Cells Using Dual Affinity Re-Targeting Molecules (DARTs®) , 2013 .

[31]  John M. Ashton,et al.  BCL-2 inhibition targets oxidative phosphorylation and selectively eradicates quiescent human leukemia stem cells. , 2013, Cell stem cell.

[32]  S. Jang,et al.  Flow cytometric quantification and immunophenotyping of leukemic stem cells in acute myeloid leukemia , 2012, Annals of Hematology.

[33]  J. Tamburini,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 , 2011, Haematologica.

[34]  J. Dick,et al.  Monoclonal antibody-mediated targeting of CD123, IL-3 receptor alpha chain, eliminates human acute myeloid leukemic stem cells. , 2009, Cell stem cell.

[35]  E. Coccia,et al.  Elevated expression of IL-3Ralpha in acute myelogenous leukemia is associated with enhanced blast proliferation, increased cellularity, and poor prognosis. , 2002, Blood.