236 The Histone Deacetylase Inhibitor SAHA Sensitizes AML Cells to a Combination of Nucleoside Analogs and the DNA-Alkylating Agent Busulfan

Track: Contributed Abstracts
Wednesday, February 13, 2013, 6:45 PM-7:45 PM
Hall 1 (Salt Palace Convention Center)
Guiyun Wang, PhD , Stem Cell Transplantation and Cellular Therapy, U. T. M. D. Anderson Cancer Center, Houston,, TX
Ben Valdez, PhD , Stem Cell Transplantation & Cellular Therapy, UT MD Anderson Cancer Center, Houston, TX
Yang Li, MS , Stem Cell Transplantation and Cellular Therapy, U. T. M. D. Anderson Cancer Center, Houston, TX
Jose R Dominguez , Stem Cell Transplantation & Cellular Therapy, UT MD Anderson Cancer Center, Houston, TX
Richard E. Champlin, MD , MD Anderson Cancer Center, Houston, TX
Borje S. Andersson, MD, PhD , Stem Cell Transplantation and Cellular Therapy, M. D. Anderson Cancer Center, Houston, TX
DNA alkylating agent busulfan (Bu) and nucleoside analogs clofarabine (Clo) and fludarabine (Flu) are commonly used as part of the pre-transplant conditioning regimen in allogeneic hematopoietic stem cell transplantation (allo-HSCT). We previously reported that a combination of [Bu+Clo+Flu] has a synergistic cytotoxicity in AML cells. We hypothesized that combination of [Bu+Clo+Flu] with histone deacetylase inhibitor SAHA will further enhance the cytotoxicity. We exposed the AML cell lines KBM3/Bu2506 (KBu) to Bu (10 μg/ml), Clo (10 nM), Flu (0.2 μM) and SAHA (0.7 μM),  and OCI-AML3 to Bu (1.5 μg/ml), Clo (10 nM), Flu (0.5 μM) and SAHA (0.7 μM) alone or in various combinations. Exposure of these cells to Bu, Clo, Flu or SAHA alone, or to  two-drug combinations [Bu+Clo], [Bu+Flu] and [Clo+Flu], did not significantly affect cell proliferation relative to the control. The combination of [Bu+Clo+Flu] resulted in 20-30% inhibition of cell proliferation. Addition of SAHA to the [Bu+Clo+Flu] mixture further enhanced the inhibition of proliferation of KBu and OCI-AML3  by 60% and 80%, respectively, suggesting synergistic cytotoxicity. Biochemical analyses suggest that this cytotoxicity may be attributed to (1) activated DNA-damage response and cell cycle checkpoint through ATM-CHK2-P53 pathway, (2) histone 3 and histone 4 modifications, and (3) activation of both the intrinsic and extrinsic pathways of apoptosis. The pro-apoptotic proteins BAX and cleaved BID increased in the mitochondria when cells were exposed to [Bu+Clo+Flu+SAHA], and further the pro-survival protein MCL-1 was cleaved and yielded a pro-apoptotic 28-kDa protein, while the level of the pro-survival protein BCL-xL decreased. Finally, phosphorylated and pan P53 increased in the mitochondria when OCI-AML3 cells that harbor wild type P53 were exposed to [Bu+Clo+Flu] or [Bu+Clo+Flu+SAHA]. These changes in the level of proteins involved in mitochondrial control of apoptosis may consequently cause mitochondrial outer membrane permeabilization (MOMP). The mitochondrial membrane potential, as a marker for MOMP, decreased by 40% or 80% when cells were exposed to [Bu+Clo+Flu] or [Bu+Clo+Flu+SAHA], respectively, and this change caused leakage of cytochrome cand SMAC/DIABLO into the cytosol leading to caspase activation, and release of apoptosis-inducing factor (AIF) into the nucleus, inducing nuclear fragmentation and cell death. These results provide a mechanistic basis for introducing SAHA as a complement to our double nucleoside-busulfan combination in pre-transplant conditioning therapy for AML patients undergoing allo-HSCT. We hypothesize that SAHA-associated enhancement of apoptosis will result in increased antitumor efficacy with retained safety in a clinical transplant program.