Leukemias affect about 60,000 individuals, and cause the death of 24,000 individuals annually in the US. The Leukemia SPORE renewal application builds upon progress achieved in the previous funding periods, which contributed to several changes in the standards of care in leukemia. It proposes five important novel mechanistic strategies which if successful, will establish new standards of therapies in leukemia: epigenetic therapy modulation; immunotherapy with a new monoclonal antibody Hu-8F4; non-genotoxic p53 modulation by MDM2 inhibition; novel natural-killer (NK) cellular therapy; targeting oxidative phosphorylation ( OxPhos) in leukemia with novel OxPhos inhibitors. Our overall goal is to discover/enhance these new therapies through a better understanding of the causal pathophysiologies in leukemia and the identification of actionable targets. We propose five fully translational research projects (laboratory to clinic and back) supported by three cores. The overall Specific Aims are: 1) Optimize and improve the efficacy of epigenetic therapies in AML (Project 1). This research area was developed by Project 1 co-leaders over the past 14 years, and resulted in the FDA approval of decitabine as an epigenetic therapy for MDS, and the European EMEA approval (2012) for the treatment of elderly AML unfit for intensive chemotherapy. The new aims investigate enhancing the epigenetic effects through suppression of CDK9. 2) Explore anti-leukemic effects of a novel targeted immune therapy using 8F4 monoclonal antibody (Project 2). Previous work through this SPORE resulted in the development of the PR1 vaccine and the discovery of a newly discovered humanized T cell receptor-like antibody (8F4) with specificity for a conformational epitope of PR1 in vitro and in vivo. . Project investigators will now test the efficacy of the new 8F4 antibody and conduct a phase I clinical trial to determine its anti- AML efficacy, and understand the mechanisms behind its success/failure in patients treated. 3) Explore strategies to enhance non-genotoxic p53 activation by MDM2 inhibition in AML (Project 3). Previous work introduced p53-targeted therapy in leukemia as promising. Investigators will now extend these findings using preclinical and clinical studies of novel MDM2 inhibitors and combinations with apoptosis inducing agents (venetoclax). 4) Investigate NK-CAR cellular therapy in leukemia (Project 4). This is an in-house therapeutic strategy funded through a Leukemia SPORE CEP and showing promising translational therapeutic value. 5) Develop OxPhos- based targeted therapies in leukemia (Project 5). This is another in-house developed approach and molecule investigated by investigators previously supported by a Leukemia SPORE DRP, and expanded into a full project based on its promising results.

Public Health Relevance

If successful, this research will legitimize novel unique mechanistic therapeutic approaches that can change the standards of care of leukemias, alone or in combination with chemotherapy: epigenetic therapy; monoclonal antibodies for acute myeloid leukemia and other leukemias; p53 modulation strategies; in-house developed natural-killer (NK) cellular therapy; and inhibition of oxidative phosphorylation ( OxPhos) with novel in-house developed OxPhos inhibitors-based combinations. The success of the SPORE will improve the prognosis and survival and cure rates in leukemia.

Agency
National Institute of Health (NIH)
Institute
National Cancer Institute (NCI)
Type
Specialized Center (P50)
Project #
5P50CA100632-17
Application #
9762850
Study Section
Special Emphasis Panel (ZCA1)
Program Officer
Kuzmin, Igor A
Project Start
2003-08-05
Project End
2023-08-31
Budget Start
2019-09-01
Budget End
2020-08-31
Support Year
17
Fiscal Year
2019
Total Cost
Indirect Cost
Name
University of Texas MD Anderson Cancer Center
Department
Internal Medicine/Medicine
Type
Hospitals
DUNS #
800772139
City
Houston
State
TX
Country
United States
Zip Code
77030
Kelly, Andrew D; Madzo, Jozef; Madireddi, Priyanka et al. (2018) Demethylator phenotypes in acute myeloid leukemia. Leukemia 32:2178-2188
Levis, Mark J; Perl, Alexander E; Altman, Jessica K et al. (2018) A next-generation sequencing-based assay for minimal residual disease assessment in AML patients with FLT3-ITD mutations. Blood Adv 2:825-831
Shah, Maitri Y; Ferracin, Manuela; Pileczki, Valentina et al. (2018) Cancer-associated rs6983267 SNP and its accompanying long noncoding RNA CCAT2 induce myeloid malignancies via unique SNP-specific RNA mutations. Genome Res 28:432-447
Masarova, Lucia; Verstovsek, Srdan; Hidalgo-Lopez, Juliana E et al. (2018) A phase 2 study of ruxolitinib in combination with azacitidine in patients with myelofibrosis. Blood 132:1664-1674
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 :

Showing the most recent 10 out of 487 publications