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 #
2P50CA100632-16
Application #
9485515
Study Section
Special Emphasis Panel (ZCA1)
Program Officer
Kuzmin, Igor A
Project Start
2003-08-05
Project End
2023-08-31
Budget Start
2018-09-01
Budget End
2019-08-31
Support Year
16
Fiscal Year
2018
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
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
Zhang, Weiguo; Ly, Charlie; Ishizawa, Jo et al. (2018) Combinatorial targeting of XPO1 and FLT3 exerts synergistic anti-leukemia effects through induction of differentiation and apoptosis in FLT3-mutated acute myeloid leukemias: from concept to clinical trial. Haematologica 103:1642-1653
Takahashi, Koichi; Wang, Feng; Morita, Kiyomi et al. (2018) Integrative genomic analysis of adult mixed phenotype acute leukemia delineates lineage associated molecular subtypes. Nat Commun 9:2670
Ishizawa, Jo; Nakamaru, Kenji; Seki, Takahiko et al. (2018) Predictive Gene Signatures Determine Tumor Sensitivity to MDM2 Inhibition. Cancer Res 78:2721-2731
Kayser, Sabine; Levis, Mark J (2018) Advances in targeted therapy for acute myeloid leukaemia. Br J Haematol 180:484-500

Showing the most recent 10 out of 487 publications