Risk, Characteristics and Biomarkers of Cytokine Release Syndrome in Patients with Relapsed/Refractory AML or MDS Treated with CD3xCD123 Bispecific Antibody APVO436

Simple Summary Cytokine release syndrome is a potentially life-threatening complication of therapy with T-cell engaging bispecific antibodies. Here we evaluated the risk, characteristics and biomarkers of treatment-emergent cytokine release syndrome in patients with relapsed/refractory acute myeloid leukemia or myelodysplastic syndrome who received weekly intravenous infusions of the CD3xCD123 bispecific antibody APVO436. Cytokine release syndrome was encountered in 10 of 46 patients (21.7%) treated with APVO436 with a cumulative Grade 3/4 cytokine release syndrome incidence of 8.7%. Cytokine profiling in patients who developed cytokine release syndrome after APVO436 infusion indicated that the predominant cytokine in this inflammatory cytokine response was IL-6. The findings from this research provide new insights regarding the biology and effective management of cytokine release syndrome in leukemia patients treated with T-cell redirecting bispecific antibodies. Abstract We evaluate the risk, characteristics and biomarkers of treatment-emergent cytokine release syndrome (CRS) in patients with relapsed/refractory acute myeloid leukemia (AML) or myelodysplastic syndrome (MDS) who received APVO436 during the dose-escalation phase of a Phase 1B study (ClinicalTrials.gov, identifier: NCT03647800). Of four patients who developed Grade ≥ 3 CRS, two received steroid prophylaxis. The dose level, gender, race, obesity, or baseline hematologic parameters in peripheral blood did not predict the risk of CRS. Patients with a higher leukemia burden as determined by a higher total WBC, higher percentage of blasts in bone marrow, or higher percentage of blasts in peripheral blood (by hematopathology or immunophenotyping) did not have a higher incidence of CRS. There was an age difference between patients who did versus patients who did not develop CRS (72.9 ± 1.6 years (Median 73.5 years) vs. 63.3 ± 2.3 years (Median: 65.0 years), which was borderline significant (p = 0.04). Premedication with steroids did not eliminate the risk of CRS. Cytokine profiling in patients who developed CRS after APVO436 infusion indicates that the predominant cytokine in this inflammatory cytokine response was IL-6. APVO436-associated CRS was generally manageable with tocilizumab with or without dexamethasone. Notably, the development of CRS after APVO436 therapy did not appear to be associated with a response. The prolonged stabilization of disease, partial remissions and complete remissions were achieved in both patients who experienced CRS, as well as patients who did not experience CRS after APVO436 infusions.

[1]  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.

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

[3]  M. Sadelain,et al.  Cytokine release syndrome and associated neurotoxicity in cancer immunotherapy , 2021, Nature Reviews Immunology.

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

[7]  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.

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

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

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

[11]  Jihye Kim,et al.  Insight into the relationship between obesity-induced low-level chronic inflammation and COVID-19 infection , 2020, International Journal of Obesity.

[12]  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.

[13]  Amber Dance What is a cytokine storm? , 2020 .

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

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

[16]  E. Elez,et al.  Cytokine release syndrome. Reviewing a new entity in the intensive care unit. , 2019, Medicina intensiva.

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

[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]  I. Aldoss,et al.  Cytokine Release Syndrome With the Novel Treatments of Acute Lymphoblastic Leukemia: Pathophysiology, Prevention, and Treatment , 2019, Current Oncology Reports.

[22]  S. Grupp,et al.  ASTCT Consensus Grading for Cytokine Release Syndrome and Neurologic Toxicity Associated with Immune Effector Cells. , 2019, Biology of blood and marrow transplantation : journal of the American Society for Blood and Marrow Transplantation.

[23]  W. Murphy,et al.  Obesity as an immune‐modifying factor in cancer immunotherapy , 2018, Journal of leukocyte biology.

[24]  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.

[25]  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.

[26]  Daniel Li,et al.  Kinetics and biomarkers of severe cytokine release syndrome after CD19 chimeric antigen receptor-modified T-cell therapy. , 2017, Blood.

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

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

[29]  J. Blankenship,et al.  MOR209/ES414, a Novel Bispecific Antibody Targeting PSMA for the Treatment of Metastatic Castration-Resistant Prostate Cancer , 2016, Molecular Cancer Therapeutics.

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

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

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

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

[34]  J. Margolick,et al.  Multisite Comparison of High-Sensitivity Multiplex Cytokine Assays , 2011, Clinical and Vaccine Immunology.

[35]  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.

[36]  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.

[37]  K. Mitani [Myelodysplastic syndrome]. , 2020, Nihon rinsho. Japanese journal of clinical medicine.

[38]  J. Dutcher,et al.  Managing toxicities of high-dose interleukin-2. , 2002, Oncology.

[39]  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.

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