Acute myelogenous leukemia (AML) is a disease in which the accumulation of primitive nonfunctional precursor cells results in the death of 80% of patients due to bleeding and infection. Although allogeneic bone marrow transplantation is curative, only 25% of AML patients are eligible for this therapy, and only half of these survive long-term. Another 10-20% of patients are cured by combination chemotherapy. In order to develop new and improved directions of therapy for AML, we are proposing to use cell cycle regulatory molecules such as growth factors or differentiation induction agents to specifically sensitize the leukemia cells to phase-specific chemotherapy agents. These treatment programs will be linked to laboratory assays which will provide short-term molecular endpoints for the evaluation of response and minimal residual disease. The molecular changes found in these leukemia cells will be used as targets for therapy as well as for predictors of response, remission duration and survival. We will carry this analysis out in untreated patients and in patients who are in remission and relapse. We will classify patients as to whether they develop a proliferative response, a differentiation induction response, or an apoptotic response to the combination of chemotherapy plus a cell cycle regulatory molecule. We will compare in vitro with in vivo responses. We will compare responses to chemotherapy and a cell cycle regulatory molecule in vitro and in vivo with respect to chemotherapy metabolism, cell cycle, cytoplasmic signal transduction molecules, growth factor synthesis, intranuclear growth regulatory proteins (1)53, WAF- 1/CIP-1, and Rb), Bcl-2/Bax and other members of the apoptosis family, and apoptotic and non-apoptotic mechanisms of cell death. We will use PCR and FISH assays specific for each of the chromosomally defined subsets of AML to define response to therapy, response to cell cycle regulatory - molecules, and minimal residual disease. We will use the information derived from these assays to allocate patients to specific programs of therapy. We will use this information to build sequential therapeutic programs which integrate chemotherapy with cell cycle regulatory molecules, used singly and in combinations. In this way, we hope to be able to develop therapy which is targeted to the molecular and biological defects in AML, which is less toxic to normal tissues and provides more durable remissions in all subsets of AML in untreated patients, in relapsed patients, and in the minimal residual disease setting. Because the principles of therapy are relevant to the treatment of epithelial neoplasms as well, we expect the information to be derived in this program to provide direction for improvements in solid tumor therapy as well.

Agency
National Institute of Health (NIH)
Institute
National Cancer Institute (NCI)
Type
Research Program Projects (P01)
Project #
5P01CA055164-09
Application #
6172629
Study Section
Subcommittee G - Education (NCI)
Program Officer
Wu, Roy S
Project Start
1992-06-01
Project End
2001-06-30
Budget Start
2000-08-04
Budget End
2001-06-30
Support Year
9
Fiscal Year
2000
Total Cost
$1,270,463
Indirect Cost
Name
University of Texas MD Anderson Cancer Center
Department
Other Basic Sciences
Type
Other Domestic Higher Education
DUNS #
001910777
City
Houston
State
TX
Country
United States
Zip Code
77030
Ruvolo, Peter P; Ruvolo, Vivian R; Burks, Jared K et al. (2018) Role of MSC-derived galectin 3 in the AML microenvironment. Biochim Biophys Acta Mol Cell Res 1865:959-969
Ngankeu, Apollinaire; Ranganathan, Parvathi; Havelange, Violaine et al. (2018) Discovery and functional implications of a miR-29b-1/miR-29a cluster polymorphism in acute myeloid leukemia. Oncotarget 9:4354-4365
Le, Phuong M; Andreeff, Michael; Battula, Venkata Lokesh (2018) Osteogenic niche in the regulation of normal hematopoiesis and leukemogenesis. Haematologica :
Jiang, Xuejie; Mak, Po Yee; Mu, Hong et al. (2018) Disruption of Wnt/?-Catenin Exerts Antileukemia Activity and Synergizes with FLT3 Inhibition in FLT3-Mutant Acute Myeloid Leukemia. Clin Cancer Res 24:2417-2429
Ishizawa, Jo; Nakamaru, Kenji; Seki, Takahiko et al. (2018) Predictive Gene Signatures Determine Tumor Sensitivity to MDM2 Inhibition. Cancer Res 78:2721-2731
Sekihara, Kazumasa; Saitoh, Kaori; Han, Lina et al. (2017) Targeting mantle cell lymphoma metabolism and survival through simultaneous blockade of mTOR and nuclear transporter exportin-1. Oncotarget 8:34552-34564
Carter, Bing Z; Mak, Po Yee; Wang, Xiangmeng et al. (2017) Focal Adhesion Kinase as a Potential Target in AML and MDS. Mol Cancer Ther 16:1133-1144
Zeng, Zhihong; Liu, Wenbin; Tsao, Twee et al. (2017) High-throughput profiling of signaling networks identifies mechanism-based combination therapy to eliminate microenvironmental resistance in acute myeloid leukemia. Haematologica 102:1537-1548
Pan, Rongqing; Ruvolo, Vivian; Mu, Hong et al. (2017) Synthetic Lethality of Combined Bcl-2 Inhibition and p53 Activation in AML: Mechanisms and Superior Antileukemic Efficacy. Cancer Cell 32:748-760.e6
Jacamo, Rodrigo; Davis, R Eric; Ling, Xiaoyang et al. (2017) Tumor Trp53 status and genotype affect the bone marrow microenvironment in acute myeloid leukemia. Oncotarget 8:83354-83369

Showing the most recent 10 out of 422 publications