A Clinical Phase 1B Study of the CD3xCD123 Bispecific Antibody APVO436 in Patients with Relapsed/Refractory Acute Myeloid Leukemia or Myelodysplastic Syndrome

Simple Summary AML is a common form of blood cancer in adults. This study was undertaken to evaluate if AML patients who have failed the available standard treatment options could tolerate and potentially benefit from a new form of therapy. This new therapy activates patients’ own immune system against AML cells. The findings from this research may provide the foundation for a potentially more effective future form of standard therapy that is less likely to fail. Abstract APVO436 is a recombinant T cell-engaging humanized bispecific antibody designed to redirect host T cell cytotoxicity in an MHC-independent manner to CD123-expressing blast cells from patients with hematologic malignancies and has exhibited single-agent anti-leukemia activity in murine xenograft models of acute myeloid leukemia (AML). In this first-in-human (FIH) multicenter phase 1B study, we sought to determine the safety and tolerability of APVO436 in R/R AML/myelodysplastic syndrome (MDS) patients and identify a clinically active recommended phase 2 dose (RP2D) level for its further clinical development. A total of 46 R/R AML/MDS patients who had failed 1–8 prior lines of therapy received APVO436 as weekly intravenous (IV) infusions at 10 different dose levels, ranging from a Minimum Anticipated Biological Effect Level (MABEL) of 0.3 mcg to 60 mcg. APVO436 exhibited a favorable safety profile with acceptable tolerability and manageable drug-related adverse events (AEs), and its maximum tolerated dose (MTD) was not reached at a weekly dose of 60 mcg. The most common APVO436-related AEs were infusion-related reactions (IRR) occurring in 13 (28.3%) patients and cytokine release syndrome (CRS) occurring in 10 (21.7%). The single dose RP2D level was identified as 0.2 mcg/kg. Preliminary efficacy signals were observed in both AML and MDS patients: Prolonged stable disease (SD), partial remissions (PR), and complete remissions (CR) were observed in R/R AML patients as best overall responses to APVO436 at the RP2D level. Three of six evaluable MDS patients had marrow CRs. The safety and preliminary evidence of efficacy of APVO436 in R/R AML and MDS patients warrant further investigation of its clinical impact potential.

[1]  F. Uckun,et al.  Risk and Severity of Cytokine Release Syndrome in Patients with Relapsed/Refractory (R/R) AML or MDS Treated with CD3xCD123 Bispecific Antibody APVO436 , 2021, Blood.

[2]  M. Heuser,et al.  Treatment for Relapsed/Refractory Acute Myeloid Leukemia , 2021, HemaSphere.

[3]  Byeong-Bae Park,et al.  Targeted Therapeutic Approach Based on Understanding of Aberrant Molecular Pathways Leading to Leukemic Proliferation in Patients with Acute Myeloid Leukemia , 2021, International journal of molecular sciences.

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

[5]  J. Bewersdorf,et al.  BiTEs, DARTS, BiKEs and TriKEs—Are Antibody Based Therapies Changing the Future Treatment of AML? , 2021, Life.

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

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

[8]  S. Jamal,et al.  Soluble interleukin-6 receptor in the COVID-19 cytokine storm syndrome , 2021, Cell Reports Medicine.

[9]  C. Craddock,et al.  Optimizing Transplant Approaches and Post-Transplant Strategies for Patients With Acute Myeloid Leukemia , 2021, Frontiers in Oncology.

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

[11]  F. Uckun,et al.  Rejuveinix Shows a Favorable Clinical Safety Profile in Human Subjects and Exhibits Potent Preclinical Protective Activity in the Lipopolysaccharide-Galactosamine Mouse Model of Acute Respiratory Distress Syndrome and Multi‐Organ Failure , 2020, Frontiers in Pharmacology.

[12]  F. Ravandi,et al.  Complete Responses in Relapsed/Refractory Acute Myeloid Leukemia (AML) Patients on a Weekly Dosing Schedule of Vibecotamab (XmAb14045), a CD123 x CD3 T Cell-Engaging Bispecific Antibody; Initial Results of a Phase 1 Study , 2020, Blood.

[13]  S. Rutella,et al.  Flotetuzumab As Salvage Therapy for Primary Induction Failure and Early Relapse Acute Myeloid Leukemia , 2020 .

[14]  T. Yau,et al.  TP53 abnormalities correlate with immune infiltration and associate with response to flotetuzumab immunotherapy in AML. , 2020, Blood advances.

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

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

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

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

[19]  P. Paschka,et al.  Updated results from phase I dose-escalation study of AMG 330, a bispecific T-cell engager molecule, in patients with relapsed/refractory acute myeloid leukemia (R/R AML). , 2020 .

[20]  M. Konopleva,et al.  A phase I/II study of IMGN632, a novel CD123-targeting antibody-drug conjugate, in patients with relapsed/refractory acute myeloid leukemia, blastic plasmacytoid dendritic cell neoplasm, and other CD123-positive hematologic malignancies. , 2020 .

[21]  H. Einsele,et al.  The BiTE (bispecific T‐cell engager) platform: Development and future potential of a targeted immuno‐oncology therapy across tumor types , 2020, Cancer.

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

[23]  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.

[24]  F. Uckun,et al.  Recurrent or Refractory High-Grade Gliomas Treated by Convection-Enhanced Delivery of a TGFβ2-Targeting RNA Therapeutic: A Post-Hoc Analysis with Long-Term Follow-Up , 2019, Cancers.

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

[26]  M. Konopleva,et al.  Clinical Profile of IMGN632, a Novel CD123-Targeting Antibody-Drug Conjugate (ADC), in Patients with Relapsed/Refractory (R/R) Acute Myeloid Leukemia (AML) or Blastic Plasmacytoid Dendritic Cell Neoplasm (BPDCN) , 2019, Blood.

[27]  R. Larson,et al.  Gilteritinib or Chemotherapy for Relapsed or Refractory FLT3-Mutated AML. , 2019, The New England journal of medicine.

[28]  Kristen L. Jones,et al.  DNA methyltransferase inhibition overcomes diphthamide pathway deficiencies underlying CD123-targeted treatment resistance. , 2019, The Journal of clinical investigation.

[29]  Catherine Lai,et al.  Recent drug approvals for acute myeloid leukemia , 2019, Journal of Hematology & Oncology.

[30]  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.

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

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

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

[34]  J. Gross,et al.  Abstract 1786: APVO436, a bispecific anti-CD123 x anti-CD3 ADAPTIR™ molecule for redirected T-cell cytotoxicity, induces potent T-cell activation, proliferation and cytotoxicity with limited cytokine release , 2018, Immunology.

[35]  W. Hiddemann,et al.  Coexpression profile of leukemic stem cell markers for combinatorial targeted therapy in AML , 2018, Leukemia.

[36]  T. Chittenden,et al.  A CD123-targeting antibody-drug conjugate, IMGN632, designed to eradicate AML while sparing normal bone marrow cells. , 2018, Blood advances.

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

[38]  J. Blankenship,et al.  Abstract 597: Bispecific anti-CD123 x anti-CD3 ADAPTIR™ molecules for redirected T-cell cytotoxicity in hematological malignancies , 2017 .

[39]  H. Kantarjian,et al.  Emerging Molecular and Immune Therapies in Acute Myeloid Leukemia , 2017 .

[40]  J. Cortes,et al.  Treatment of Relapsed/Refractory Acute Myeloid Leukemia , 2017, Current Treatment Options in Oncology.

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

[42]  Shery Jacob,et al.  A simple practice guide for dose conversion between animals and human , 2016, Journal of basic and clinical pharmacy.

[43]  M. Weiss,et al.  Immunotherapy in acute myeloid leukemia , 2015, Cancer.

[44]  C. Sentman,et al.  Bispecific T‐cell engagers for cancer immunotherapy , 2015, Immunology and cell biology.

[45]  S. Grupp,et al.  Current concepts in the diagnosis and management of cytokine release syndrome. , 2014, Blood.

[46]  U. Testa,et al.  CD 123 is a membrane biomarker and a therapeutic target in hematologic malignancies , 2014, Biomarker Research.

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

[48]  R. Solana,et al.  Cytokine profiles in acute myeloid leukemia patients at diagnosis: survival is inversely correlated with IL-6 and directly correlated with IL-10 levels. , 2013, Cytokine.

[49]  M. Boyiadzis,et al.  CD123 Immunohistochemical Expression in Acute Myeloid Leukemia is Associated With Underlying FLT3-ITD and NPM1 Mutations , 2012, Applied immunohistochemistry & molecular morphology : AIMM.

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

[51]  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.

[52]  F. Lo‐Coco,et al.  Immunophenotypic features of acute myeloid leukaemia patients exhibiting high FLT3 expression not associated with mutations , 2011, British journal of haematology.

[53]  Peter Lloyd,et al.  The minimum anticipated biological effect level (MABEL) for selection of first human dose in clinical trials with monoclonal antibodies. , 2009, Current opinion in biotechnology.

[54]  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.

[55]  R. Foà,et al.  Interleukin (IL)‐3/granulocyte macrophage‐colony stimulating factor/IL‐5 receptor alpha and beta chains are preferentially expressed in acute myeloid leukaemias with mutated FMS‐related tyrosine kinase 3 receptor , 2009, British journal of haematology.

[56]  H. Einsele,et al.  Tumor Regression in Cancer Patients by Very Low Doses of a T Cell–Engaging Antibody , 2008, Science.

[57]  A. Frankel,et al.  Phase I clinical study of diphtheria toxin-interleukin 3 fusion protein in patients with acute myeloid leukemia and myelodysplasia , 2008, Leukemia & lymphoma.

[58]  T. Lister,et al.  Hematopoietic stem cells express multiple myeloid markers: implications for the origin and targeted therapy of acute myeloid leukemia. , 2005, Blood.

[59]  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.

[60]  Irving L. Weissman,et al.  Prospective isolation of human clonogenic common myeloid progenitors , 2002, Proceedings of the National Academy of Sciences of the United States of America.

[61]  D. Howard,et al.  The interleukin-3 receptor alpha chain is a unique marker for human acute myelogenous leukemia stem cells , 2000, Leukemia.

[62]  A. Ho,et al.  Correlation Between IL‐3 Receptor Expression and Growth Potential of Human CD34+ Hematopoietic Cells from Different Tissues , 1999, Stem cells.

[63]  G. Wagemaker,et al.  Differential expression of receptors for hemopoietic growth factors on subsets of CD34+ hemopoietic cells. , 1996, Leukemia & lymphoma.

[64]  K. Arai,et al.  Expression and factor-dependent modulation of the interleukin-3 receptor subunits on human hematopoietic cells. , 1993, Blood.