Our long-term goal Is to develop Immune therapies that target aberrantly expressed proteases In blasts and leukemia stem cells. PR1 peptide (VLQELNVTV) is a peptide derived from the leukemia- associated antigens proteinase 3 (P3) and neutrophil elastase (NE), which is presented on HLA-A2 to PR1- speclfic cytotoxic T lymphocytes (PR1-CTL). During the last grant period, we showed that PR1 is cross- presented by dendritic cells (DCs) and B cells, and we showed the mechanism required proteasome cleavage exogenous P3 and NE taken up by antigen-presenting cells. We completed a phase 2 PRI peptide vaccine trial that showed immune responses in 58% and clinical responses in 16%, but responses were limited to patients with low leukemia burden. We developed a strategy to adoptively transfer donor- derived PR1-CTL to stem cell transplant recipients to treat high leukemia burden. As an alternate strategy, we identified a T cell receptor (TCR)-like monoclonal antibody (8F4) with specificity for a conformational epitope of PR1 bound to the HLA-A2 molecule. 8F4 induces complement-dependent cytotoxicity (CDC) of AML and LSC and inhibits AML progenitor cell growth but not normal bone marrow progenitors, which supports further study of 8F4 as a potential therapeutic monoclonal antibody (mAb) for AML. Our preliminary data show that 8F4 prevents AML engraftment and reduced or eliminated established primary human AML xenografts in vivo. 8F4 also significantly reduced or eliminated leukemia stem cells (LSCs) in mouse models. We produced a humanized 8F4 (Hu8F4) and showed it significantly reduced (>95%) established human AML xenografts. Thus, 8F4 is the first TCR-like mAb that inhibits growth of AML and LSC and eliminates AML in vivo. In prior years of this grant, we have used a PRI peptide vaccine to induce immunity in patients with AML, CML, and MDS and more recently we initiated two adoptive cell therapy trials with PRI-specific CTL in AML and CML, respectively. In this proposal, we will characterize the 8F4 mAb as a novel therapeutic modality for leukemia. Our central hypothesis is that immune therapy targeting surface PR1/HLA-A2 molecules can selectively eliminate leulfemia and leuliemia stem cells, but not normal hematopoietic cells. To investigate the therapeutic potential of this novel antibody to improve the outcome of patients with myeloid malignancies, we will pursue the following Specific Aims 1) characterize the activity of HU-8F4 against human AML, 2) perform pre-clinical treatment validation and safety studies of Hu-8F4, and 3) conduct a first-in-human phase 1 clinical trial with Hu8F4 in patients with relapsed AML.

Public Health Relevance

Successful completion of these studies will provide the biological, pre-clinical and clinical bases for exploring the therapeutic potential of a highly novel TCR-like antibody (8F4) to treat leukemia. 8F4 eliminates leukemia and leukemia stem cells in mouse models of primary human leukemia without eliminating normal blood cells or normal hematopoietic stem cells. Therefore, 8F4 shows promise as a highly AML-specific agent with low potential for toxicity. We will conduct a first-in-human clinical trial with this antibody in AML.

Agency
National Institute of Health (NIH)
Institute
National Cancer Institute (NCI)
Type
Specialized Center (P50)
Project #
2P50CA100632-11
Application #
8499745
Study Section
Special Emphasis Panel (ZCA1-RPRB-7 (J1))
Project Start
2013-05-01
Project End
2018-08-31
Budget Start
2013-09-13
Budget End
2014-08-31
Support Year
11
Fiscal Year
2013
Total Cost
$196,339
Indirect Cost
Name
University of Texas MD Anderson Cancer Center
Department
Type
DUNS #
800772139
City
Houston
State
TX
Country
United States
Zip Code
77030
Good, Charly Ryan; Panjarian, Shoghag; Kelly, Andrew D et al. (2018) TET1-Mediated Hypomethylation Activates Oncogenic Signaling in Triple-Negative Breast Cancer. Cancer Res 78:4126-4137
Choi, Sangbum; Kang, Sangwook; Huang, Xuelin (2018) Smoothed quantile regression analysis of competing risks. Biom J 60:934-946
Boddu, Prajwal; Kantarjian, Hagop; Garcia-Manero, Guillermo et al. (2018) The emerging role of immune checkpoint based approaches in AML and MDS. Leuk Lymphoma 59:790-802
Yang, Tian-Hui; St John, Lisa S; Garber, Haven R et al. (2018) Membrane-Associated Proteinase 3 on Granulocytes and Acute Myeloid Leukemia Inhibits T Cell Proliferation. J Immunol 201:1389-1399
Rivera-Del Valle, Nilsa; Cheng, Tiewei; Irwin, Mary E et al. (2018) Combinatorial effects of histone deacetylase inhibitors (HDACi), vorinostat and entinostat, and adaphostin are characterized by distinct redox alterations. Cancer Chemother Pharmacol 81:483-495
Le, Phuong M; Andreeff, Michael; Battula, Venkata Lokesh (2018) Osteogenic niche in the regulation of normal hematopoiesis and leukemogenesis. Haematologica :
Zhang, Hanghang; Pandey, Somnath; Travers, Meghan et al. (2018) Targeting CDK9 Reactivates Epigenetically Silenced Genes in Cancer. Cell 175:1244-1258.e26
Morita, Kiyomi; Kantarjian, Hagop M; Wang, Feng et al. (2018) Clearance of Somatic Mutations at Remission and the Risk of Relapse in Acute Myeloid Leukemia. J Clin Oncol 36:1788-1797
Fiorini, Elena; Santoni, Andrea; Colla, Simona (2018) Dysfunctional telomeres and hematological disorders. Differentiation 100:1-11
Cortes, Jorge; Perl, Alexander E; Döhner, Hartmut et al. (2018) Quizartinib, an FLT3 inhibitor, as monotherapy in patients with relapsed or refractory acute myeloid leukaemia: an open-label, multicentre, single-arm, phase 2 trial. Lancet Oncol 19:889-903

Showing the most recent 10 out of 487 publications