The long-term goal of this project is to develop and translate into early phase clinical trials novel antibody-based reagents for the treatment of acute myelogenous leukemia (AML). Less than half of AML patients are cured with current treatment approaches, and relapse and refractory AML patients have a median overall survival of about 4 months. Allogeneic hematopoietic cell transplant (alloHCT) remains the only curative therapy for patients with relapsed and refractory AML. However, wider application of alloHCT has been limited by the morbidity associated with the procedure, which arises from graft-versus-host disease and from the toxicity of the conditioning regimen, a course of cytotoxic drugs and/or irradiation that is used to enable engraftment of donor hematopoietic stem and progenitor cells (HSPCs) in the bone marrow. In this proposal, we will utilize antibody-based immunotherapy approaches to treat AML relapse and allow for alloHCT without the need for chemotherapy and/or irradiation.
In Aim 1, we will conduct a ?first-in-human? Phase I clinical trial of AMV564, a CD33 x CD3 tetravalent bispecific antibody in patients with relapsed or refractory AML (ClinicalTrials.gov id: NCT03144245). This study is designed in two segments: a Dose Escalation Stage testing up to 10 doses of AMV564 at 3 patients per dose, followed by an Expansion Stage segment at the recommended Phase 2 dose of AMV564. We will characterize the immunomodulatory activity and potential anti-tumor activity of AMV564 by completing correlative studies evaluating (1) serum cytokines, (2) AML and T cell subset numbers, phenotype and function and (3) the subclonal architecture of AML blasts.
In Aim 2, we will determine if antibody-based drug conjugates can mitigate cytokine release syndrome (CRS) associated with T cell immunotherapeutics, kill AML blasts and provide conditioning for alloHSCT. T cell engaging therapies, which include bispecific retargeting reagents like AMV564, have been limited by CRS. We will test if targeted depletion of macrophage and monocyte populations with toxin-conjugated antibody fragments or modulation of the IFN? and IL-6 signaling pathways will prevent CRS mediated by T cell engaging therapies. If successful, these CRS-mitigating strategies would reduce the main limitation currently facing T cell engaging therapies. Next, we will test if toxin-conjugated antibody antigen binding fragments (Fabs) or bispecific antibodies targeting HSPCs/AML (via CD45 or c-Kit) and T lymphocytes/NK cells (via CD2 or CD7) can simultaneously treat AML and provide conditioning for alloHCT without the need for radiation or chemotherapy. The potential applications of this more targeted and less toxic conditioning regimen are not limited to alloHCT alone, but would also provide a platform for autologous gene therapy for inborn errors of metabolism, hemoglobinopathies, and primary immune deficiencies.

Public Health Relevance

Most patients with acute myeloid leukemia (AML) die from progressive disease after relapse. In this project we will perform a first-in-human clinical trial testing a novel immunotherapeutic reagent for the treatment of AML. We will also develop novel treatment regimens to (1) mitigate cytokine release syndrome after administration of T cell engaging therapies and (2) simultaneously kill AML blasts and condition recipients for allogeneic hematopoietic cell transplant by ablating host hematopoietic stem and immune cells without the need for toxic chemotherapy and/or irradiation.

Agency
National Institute of Health (NIH)
Institute
National Cancer Institute (NCI)
Type
Specialized Center (P50)
Project #
5P50CA171963-07
Application #
9756324
Study Section
Special Emphasis Panel (ZCA1)
Project Start
Project End
Budget Start
2019-07-01
Budget End
2020-06-30
Support Year
7
Fiscal Year
2019
Total Cost
Indirect Cost
Name
Washington University
Department
Type
DUNS #
068552207
City
Saint Louis
State
MO
Country
United States
Zip Code
63130
Duncavage, Eric J; Jacoby, Meagan A; Chang, Gue Su et al. (2018) Mutation Clearance after Transplantation for Myelodysplastic Syndrome. N Engl J Med 379:1028-1041
Khoury, Hanna Jean; Langston, Amelia A; Kota, Vamsi K et al. (2018) Ruxolitinib: a steroid sparing agent in chronic graft-versus-host disease. Bone Marrow Transplant 53:826-831
Schroeder, Mark A; Choi, Jaebok; Staser, Karl et al. (2018) The Role of Janus Kinase Signaling in Graft-Versus-Host Disease and Graft Versus Leukemia. Biol Blood Marrow Transplant 24:1125-1134
Perry, Justin S A; Russler-Germain, Emilie V; Zhou, You W et al. (2018) CD36 Mediates Cell-Surface Antigens to Promote Thymic Development of the Regulatory T Cell Receptor Repertoire and Allo-tolerance. Immunity 48:923-936.e4
Wong, Terrence N; Miller, Christopher A; Jotte, Matthew R M et al. (2018) Cellular stressors contribute to the expansion of hematopoietic clones of varying leukemic potential. Nat Commun 9:455
Christopher, Matthew J; Petti, Allegra A; Rettig, Michael P et al. (2018) Immune Escape of Relapsed AML Cells after Allogeneic Transplantation. N Engl J Med 379:2330-2341
Trissal, Maria C; Wong, Terrence N; Yao, Juo-Chin et al. (2018) MIR142 Loss-of-Function Mutations Derepress ASH1L to Increase HOXA Gene Expression and Promote Leukemogenesis. Cancer Res 78:3510-3521
Jacoby, Meagan A; Duncavage, Eric J; Chang, Gue Su et al. (2018) Subclones dominate at MDS progression following allogeneic hematopoietic cell transplant. JCI Insight 3:
Monlish, Darlene A; Bhatt, Sima T; Duncavage, Eric J et al. (2018) Loss of Toll-like receptor 2 results in accelerated leukemogenesis in the NUP98-HOXD13 mouse model of MDS. Blood 131:1032-1035
Choi, Jaebok; Cooper, Matthew L; Staser, Karl et al. (2018) Baricitinib-induced blockade of interferon gamma receptor and interleukin-6 receptor for the prevention and treatment of graft-versus-host disease. Leukemia 32:2483-2494

Showing the most recent 10 out of 64 publications