Purpose: To determine the possibility of synergistic antileukemic activity and the underlying molecular mechanisms associated with cytarabine combined with valproic acid (VPA; a histone deacetylase inhibitor and a Food and Drug Administration–licensed drug for treating both children and adults with epilepsy) in pediatric acute myeloid leukemia (AML).

Experimental Design: The type and extent of antileukemic interactions between cytarabine and VPA in clinically relevant pediatric AML cell lines and diagnostic blasts from children with AML were determined by MTT assays and standard isobologram analyses. The effects of cytarabine and VPA on apoptosis and cell cycle distributions were determined by flow cytometry analysis and caspase enzymatic assays. The effects of the two agents on DNA damage and Bcl-2 family proteins were determined by Western blotting.

Results: We showed synergistic antileukemic activities between cytarabine and VPA in four pediatric AML cell lines and nine diagnostic AML blast samples. t(8;21) AML blasts were significantly more sensitive to VPA and showed far greater sensitivities to combined cytarabine and VPA than non-t(8;21) AML cases. Cytarabine and VPA cooperatively induced DNA double-strand breaks, reflected in induction of γH2AX and apoptosis, accompanied by activation of caspase-9 and caspase-3. Further, VPA induced Bim expression and short hairpin RNA knockdown of Bim resulted in significantly decreased apoptosis induced by cytarabine and by cytarabine plus VPA.

Conclusions: Our results establish global synergistic antileukemic activity of combined VPA and cytarabine in pediatric AML and provide compelling evidence to support the use of VPA in the treatment of children with this deadly disease. Clin Cancer Res; 16(22); 5499–510. ©2010 AACR.

Translational Relevance

In this study, we show highly synergistic antileukemic activities of combined cytarabine [1-β-d-arabinofuranosylcytosine (ara-C)] and valproic acid (VPA) in a panel of pediatric acute myeloid leukemia (AML) cell lines and diagnostic blast samples derived from children with de novo AML. Thus, VPA could be an attractive agent for combination therapy for children with this deadly disease. Based on our results, VPA was recently incorporated into one of the treatment arms for high-risk AML in the St. Jude Children's Research Hospital AML08 clinical trial “A Randomized Trial of Clofarabine Plus Cytarabine Versus Conventional Induction Therapy and of Natural Killer Cell Transplantation Versus Conventional Consolidation Therapy in Patients with Newly Diagnosed Acute Myeloid Leukemia.” In this trial, children with acute megakaryocytic leukemia without t(1;22) and other high-risk patients without FLT3-ITD will receive a combination of VPA with low-dose ara-C, daunorubicin, and etoposide (LD-ADE) during the second induction course.

Acute myeloid leukemia (AML) accounts for one fourth of acute leukemias in children, but it is responsible for more than half of the leukemia deaths in this patient population (1). In contrast to the tremendous success in treating acute lymphoblastic leukemia over the last 3 decades, resulting in a >80% cure rate, improvements in AML therapy have been limited (1). Resistance to cytarabine, the most active drug in the treatment of AML, is a major cause of treatment failure (2, 3). Therefore, new therapies for children with AML are urgently needed.

Cytarabine is a prodrug that must be converted to a triphosphate derivative [1-β-d-arabinofuranosylcytosine 5′-triphosphate (ara-CTP)] to exert its cytotoxic effects (4). Cytarabine cytotoxicity is believed to result from a combination of DNA polymerase inhibition and incorporation of ara-CTP into DNA, resulting in chain termination and a blockade of DNA synthesis (4). In addition, previous studies have documented the ability of cytarabine to trigger apoptosis in human leukemia cells (4).

Histone deacetylase (HDAC) inhibitors (HDACI) promote histone acetylation and subsequent chromatin relaxation and uncoiling, which facilitates transcription of different genes, especially those involved in cellular differentiation (5). HDACIs may also disrupt the function of HDACs in corepressor complexes implicated in the differentiation blockade exhibited by certain forms of AML [e.g., t(8;21) AML and APL involving t(15;17); refs. 6, 7]. HDACI cytotoxicity is regulated by diverse mechanisms including activation of stress-related pathways or inactivation of cytoprotective pathways, upregulation of death receptors, induction of p21CIP1, ceramide production, disruption of heat shock proteins, and induction of oxidative damage (8). Further, emerging evidence suggests that HDACIs can directly induce DNA damage in leukemia cells (9). Several HDACIs are currently being tested in clinical trials, and encouraging results have been reported for their use in treating both hematologic malignancies and solid tumors (1016). However, no HDACIs have yet been approved by the U.S. Food and Drug Administration for treating children with cancer.

Recently, the anticonvulsant drug valproic acid (VPA) was reported to exhibit powerful HDACI activity (17, 18) and to induce apoptosis in leukemia cells but not in normal cells at clinically achievable concentrations (100-150 μg/mL; refs. 1820). VPA is usually well tolerated in children, and the extensive clinical experience with this drug makes it a very attractive agent for treating pediatric AML. In fact, preclinical and clinical studies have shown additive-to-synergistic antileukemic effects on AML when VPA is used in combination with other chemotherapy agents including idarubicin (21), 5-aza-2′-deoxycytidine (22), gemtuzumab ozogamicin (23), and NPI-0052 (24). Recently, VPA was reported to markedly increase cytarabine cytotoxicity in a single AML cell line (25). However, neither the mechanisms of interaction between VPA and cytarabine nor the extent to which these results can be generalized to different AML subtypes has been established.

In this study, we hypothesize that VPA synergizes with cytarabine, resulting in enhanced antileukemic activity in AML cells, by inducing apoptosis. To model this concept, we examined the effect of VPA on cytarabine cytotoxicities in four pediatric AML cell lines and nine diagnostic blast samples from children with de novo AML. We show highly synergistic antileukemic activities of combined cytarabine and VPA in all of the cell lines and diagnostic blast samples, especially those with t(8;21). Our mechanistic studies reveal cooperative induction of DNA damage by cytarabine and VPA and induction of Bim by VPA that underlie the synergistic activity of this drug combination. Collectively, our results provide compelling evidence to support the use of VPA in combination with standard chemotherapy drugs in clinical trials for treating pediatric AML.

Clinical samples

Diagnostic bone marrow samples (n = 9) from children with de novo AML were obtained from the Children's Hospital of Michigan leukemia cell bank. Cell bank samples were selected from cases with sufficient cell numbers (minimum 5 × 106, blast percentage >75%, viability >85%). Patient characteristics are summarized in Supplementary Table S1. Mononuclear cells were purified by standard Ficoll-Hypaque density centrifugation. Informed consent was provided according to the Declaration of Helsinki. Sample handling and data analysis protocols were approved by the Human Investigation Committee of the Wayne State University School of Medicine.

Drugs

Cytarabine and VPA were purchased from Sigma Chemical Co.

Cell culture

The THP-1 [derived from a 1-year-old male with AML M5 and t(9;11)], Kasumi-1 [derived from a 7-year-old male with AML M2 and t(8;21)], and MV4-11 [derived from a 10-year-old male with AML M5 and t(4;11)] pediatric AML cell lines were purchased from the American Type Culture Collection. The CMS (derived from a 2-year-old female with AML M7) pediatric AML cell line was a gift from Dr. A. Fuse (National Institute of Infectious Diseases, Tokyo, Japan). These cell lines were cultured in RPMI 1640 with 10% to 20% fetal bovine serum (FBS; Hyclone) and 2 mmol/L l-glutamine, plus 100 units/mL penicillin and 100 μg/mL streptomycin, in a 37°C humidified atmosphere containing 5% CO2/95% air.

In vitro cytotoxicity assays

In vitro cytarabine and VPA cytotoxicities of pediatric AML cell lines and diagnostic blasts were measured by using MTT (Sigma) assays, as previously described (26). IC50 values were calculated as drug concentrations necessary to inhibit 50% proliferation compared with untreated control cells. The extent and direction of cytarabine and VPA cytotoxic interactions were evaluated as described previously (27, 28). Briefly, synergism, additivity, or antagonism was quantified by determining the combination index (CI), where CI < 1, CI = 1, and CI > 1 indicate synergistic, additive, and antagonistic effects, respectively. Based on the classic isobologram, the CI was calculated using the following equation: CI = [(D)1/(Dx)1] + [(D)2/(Dx)2]. At the 50% inhibition level, (Dx)1 and (Dx)2 are concentrations of cytarabine and VPA, respectively, which induce a 50% inhibition in cell proliferation when administered individually. (D)1 and (D)2 are concentrations of cytarabine and VPA, respectively, which inhibit cell proliferation by 50% when combined.

Assessment of baseline and drug-induced apoptosis

Diagnostic AML blasts from patient 7 [46, XY, t(8;21)], THP-1, and Kasumi-1 cells cultured in RPMI 1640 plus 10% to 20% FBS were treated with VPA (0.5, 0.66, and 0.5 mmol/L, respectively) or cytarabine (1,000, 900, and 100 nmol/L, respectively) alone or in combination for 24 hours (for the patient sample) or 96 hours (for the cell lines). The VPA and cytarabine doses for the cell lines were IC20s, whereas those for patient AML blasts were ∼IC50s, determined by MTT assays. The same concentrations of VPA and cytarabine were used in the rest of the studies unless specified. The cells were harvested and vigorously pipetted, and triplicate samples were taken to determine baseline and drug-induced apoptosis using the Apoptosis Annexin V–FITC/Propidium Iodide (PI) kit (Beckman Coulter), as previously described (29). Apoptotic events were recorded as a combination of Annexin V+/PI (early apoptotic) and Annexin V+/PI+ (late apoptotic/dead) events, and results were expressed as percent of Annexin V+ cells after subtracting results for untreated cells. Synergy was quantified using the cooperativity index (cooperativity index = sum of apoptosis of single-agent treatment/apoptosis on combined treatment). Cooperativity index <1, 1, or >1 is termed synergistic, additive, or antagonistic, respectively (23).

Effects of VPA and cytarabine on cell cycle progression in AML cells

THP-1 or Kasumi-1 cells were treated with VPA or cytarabine alone or in combination for 96 hours. Cells were harvested and fixed with ice-cold 70% (v/v) ethanol for 24 hours. After centrifugation at 200 × g for 5 minutes, the cell pellets were washed with PBS (pH 7.4) and resuspended in PBS containing PI (50 μg/mL), Triton X-100 (0.1%, v/v), and DNase-free RNase (1 μg/mL). DNA contents were determined by flow cytometry using a FACScan flow cytometer (BD Biosciences). Cell cycle analysis was done with the Multicycle software (Phoenix Flow Systems, Inc.).

Western blot analysis

Extracted or immunoprecipitated proteins were subjected to SDS-PAGE. Separated proteins were electrophoretically transferred to polyvinylidene difluoride membranes (Thermo Fisher, Inc.) and immunoblotted with anti–acetyl-histone 3 (ac-H3), anti–ac-H4, anti-H4 (Upstate Biotechnology), anti-Bak, anti-Bax, anti-Bid, anti-Bim, anti-Bad, anti-Puma, anti-p21, anti–Bcl-2, anti–Bcl-xL, anti–Mcl-1, anti-γH2AX (Cell Signaling Technology), or anti–β-actin (Sigma) antibody, as described previously (30). Immunoreactive proteins were visualized using the Odyssey Infrared Imaging System (Li-COR), as described by the manufacturer.

Caspase-9 and caspase-3 assays

THP-1 and Kasumi-1 cells were treated with cytarabine or VPA alone or combined for up to 96 hours. Caspase-3 and caspase-9 enzymatic activities were assayed using the Caspase-3 Fluorometric kit and the Caspase-9 Colorimetric kit, respectively, purchased from R&D Systems, based on the manufacturer's instructions. THP-1 and Kasumi-1 cells treated with 500 and 1,000 nmol/L daunorubicin, respectively, for 16 hours (results in >70% apoptosis) were used as positive controls.

Short hairpin RNA knockdown of Bim in THP-1 cells

Bim short hairpin RNA (shRNA) lentivirus clones were purchased from the RNAi Consortium (Sigma-Aldrich). THP-1 cells were infected by shRNA lentivirus clones. After selection with puromycin, a pool of infected cells was expanded and tested for Bim expression by Western blotting (designated Bim-shRNA). A pool of cells from the negative control transduction was used as the negative control (designated NTC-shRNA).

Statistical analysis

Differences in cytarabine IC50s between VPA-treated and untreated AML cells and differences in cell apoptosis between cytarabine and VPA-treated (individually or combined) and untreated cells were compared using the paired t test. The relationship between the levels of γH2AX and caspase-3 activities was determined by the Pearson test. Statistical analyses were done with GraphPad Prism 4.0.

Synergistic antileukemic interactions between cytarabine and VPA in pediatric AML cell lines and diagnostic blasts

To explore the possibility of synergistic cytotoxicity when cytarabine was combined with HDACIs to treat pediatric AMLs, we tested VPA [a short-chain fatty acid HDACI that inhibits class I and IIa HDACs (5)] with cytarabine toward THP-1 AML cells using MTT assays. In vitro incubation of THP-1 cells with VPA alone resulted in inhibition of cell proliferation with an IC50 of 2.97 mmol/L (Fig. 1A). This was accompanied by hyperacetylation of histones H3 and H4, but not total histone H4 (Fig. 1B). This VPA concentration was in excess of the maximally achievable plasma concentration in children (1 mmol/L), at which there was only modest inhibition of cell proliferation (Fig. 1A). When simultaneously administered with cytarabine, VPA at 0.5 and 1 mmol/L significantly enhanced cytarabine sensitivity [as reflected in decreased IC50s] by 2.1- and 4.3-fold, respectively (Fig. 1C). The combined effects of cytarabine with VPA on cell proliferation were clearly synergistic, as determined by standard isobologram analysis (Fig. 1D) and by calculating CI values (28). A CI < 1, indicative of synergism, was calculated for each of the drug combinations (Table 1).

Fig. 1.

Synergistic cytotoxic interactions between VPA and cytarabine toward THP-1 cells. A, THP-1 cells were cultured at 37°C for 96 h in complete medium with dialyzed FBS in 96-well plates at a density of 4 × 104 cells/mL, with a range of concentrations of VPA, and viable cell numbers were determined using the MTT reagent and a visible microplate reader. The IC50 values were calculated as the concentrations of drug necessary to inhibit 50% proliferation compared with control cells cultured in the absence of drug. Columns, mean of at least three independent experiments; bars, SE. B, THP-1 cells were harvested and lysed after incubation with a range of concentrations of VPA (0-8 mmol/L) for 48 h. Soluble proteins were analyzed on Western blots probed by anti–ac-H3, anti–ac-H4, or anti-H4 antibody. C, cytarabine IC50s of THP-1 cells were determined in the presence or absence of VPA treated simultaneously. **, P < 0.005. D, standard isobologram analysis of THP-1 cell proliferation inhibition by VPA and cytarabine. The IC50 values of each drug are plotted on the axes; the solid line represents the additive effect, whereas the points represent the concentrations of each drug resulting in 50% inhibition of proliferation. Points falling below the line indicate synergism between drug combinations, whereas those falling above the line indicate antagonism. E, in vitro VPA sensitivities of the diagnostic AML blasts were measured by MTT assay, as described in Materials and Methods. The horizontal lines indicate median VPA IC50s in each group of patient samples. The P value was determined by the nonparametric Mann-Whitney U test. F, fold decrease of cytarabine IC50s for the diagnostic AML blasts measured by MTT assays in the presence of 0.5 mmol/L or lower VPA compared with that from cytarabine alone. The horizontal lines indicate the median fold change in each group of patient samples. The P value was determined by the nonparametric Mann-Whitney U test.

Fig. 1.

Synergistic cytotoxic interactions between VPA and cytarabine toward THP-1 cells. A, THP-1 cells were cultured at 37°C for 96 h in complete medium with dialyzed FBS in 96-well plates at a density of 4 × 104 cells/mL, with a range of concentrations of VPA, and viable cell numbers were determined using the MTT reagent and a visible microplate reader. The IC50 values were calculated as the concentrations of drug necessary to inhibit 50% proliferation compared with control cells cultured in the absence of drug. Columns, mean of at least three independent experiments; bars, SE. B, THP-1 cells were harvested and lysed after incubation with a range of concentrations of VPA (0-8 mmol/L) for 48 h. Soluble proteins were analyzed on Western blots probed by anti–ac-H3, anti–ac-H4, or anti-H4 antibody. C, cytarabine IC50s of THP-1 cells were determined in the presence or absence of VPA treated simultaneously. **, P < 0.005. D, standard isobologram analysis of THP-1 cell proliferation inhibition by VPA and cytarabine. The IC50 values of each drug are plotted on the axes; the solid line represents the additive effect, whereas the points represent the concentrations of each drug resulting in 50% inhibition of proliferation. Points falling below the line indicate synergism between drug combinations, whereas those falling above the line indicate antagonism. E, in vitro VPA sensitivities of the diagnostic AML blasts were measured by MTT assay, as described in Materials and Methods. The horizontal lines indicate median VPA IC50s in each group of patient samples. The P value was determined by the nonparametric Mann-Whitney U test. F, fold decrease of cytarabine IC50s for the diagnostic AML blasts measured by MTT assays in the presence of 0.5 mmol/L or lower VPA compared with that from cytarabine alone. The horizontal lines indicate the median fold change in each group of patient samples. The P value was determined by the nonparametric Mann-Whitney U test.

Close modal
Table 1.

Effect of VPA on cytarabine sensitivity in AML cell lines and primary AML blasts

Cell line/patientCytogeneticsVPA IC50 (mmol/L)Cytarabine IC50 (nmol/L)P
0.0 mmol/L VPA0.15 mmol/L VPA0.30 mmol/L VPA0.50 mmol/L VPA1.0 mmol/L VPA
Kasumi-1 45<2n> -X, t(8;21), complex karyotype 0.79 ± 0.03 436.3 ± 41.9 144.7 ± 35.3 (0.522) 52.1 ± 15.4 (0.499) 26.2 ± 1.6 (0.693) ND <0.004 
CMS 46, complex karyotype 2.70 ± 0.16 253.5 ± 7.7 ND ND 132.6 ± 2.7 (0.705) 125.0 ± 6.9 (0.863) <0.004 
MV4-11 48 (46-48) <2n>XY, t(4;11), complex karyotype 0.37 ± 0.03 106.3 ± 63 23.2 ± 3.4 (0.759) 3.1 ± 0.9 (0.840) ND ND <0.013 
THP-1 94 (88-96) <4n> XY/XXY, t(9;11), complex karyotype 2.97 ± 0.10 3,328.5 ± 258.4 ND ND 1,567.3 ± 134.0 (0.641) 775.3 ± 62.1 (0.574) <0.002 
Patient 1 46, XX 1.09 14,164.0 ND ND 1,086.0 (0.536) 88.0 (0.923) NA 
Patient 2 46, XY, inv(16) 4.89 6,692.0 ND ND 5,072.0 (0.867) 2,645.0 (0.599) NA 
Patient 3 46, XY, inv(16) 2.04 3,848.0 ND ND 2,476.0 (0.888) 1,552.0 (0.893) NA 
Patient 4 46, XY 1.73 2,282.0 ND ND 1,578.0 (0.980) 491.0 (0.793) NA 
Patient 5 46, XY, t(3;5) 0.91 2,191.0 ND ND 578.0 (0.812) ND NA 
Patient 6 46, XY, +9 1.04 440.3 ND ND 175.2 (0.879) ND NA 
Patient 7 46, XY, t(8;21) 0.74 902.4 ND ND 138.5 (0.825) ND NA 
Patient 8 46, XX, t(8;21) 0.72 2,228.0 306.1 (0.346) 89.51 (0.459) 34.73 (0.714) ND NA 
Patient 9 46, XX, t(8;21) 0.18 495.9 9.177 (0.861) ND ND ND NA 
Cell line/patientCytogeneticsVPA IC50 (mmol/L)Cytarabine IC50 (nmol/L)P
0.0 mmol/L VPA0.15 mmol/L VPA0.30 mmol/L VPA0.50 mmol/L VPA1.0 mmol/L VPA
Kasumi-1 45<2n> -X, t(8;21), complex karyotype 0.79 ± 0.03 436.3 ± 41.9 144.7 ± 35.3 (0.522) 52.1 ± 15.4 (0.499) 26.2 ± 1.6 (0.693) ND <0.004 
CMS 46, complex karyotype 2.70 ± 0.16 253.5 ± 7.7 ND ND 132.6 ± 2.7 (0.705) 125.0 ± 6.9 (0.863) <0.004 
MV4-11 48 (46-48) <2n>XY, t(4;11), complex karyotype 0.37 ± 0.03 106.3 ± 63 23.2 ± 3.4 (0.759) 3.1 ± 0.9 (0.840) ND ND <0.013 
THP-1 94 (88-96) <4n> XY/XXY, t(9;11), complex karyotype 2.97 ± 0.10 3,328.5 ± 258.4 ND ND 1,567.3 ± 134.0 (0.641) 775.3 ± 62.1 (0.574) <0.002 
Patient 1 46, XX 1.09 14,164.0 ND ND 1,086.0 (0.536) 88.0 (0.923) NA 
Patient 2 46, XY, inv(16) 4.89 6,692.0 ND ND 5,072.0 (0.867) 2,645.0 (0.599) NA 
Patient 3 46, XY, inv(16) 2.04 3,848.0 ND ND 2,476.0 (0.888) 1,552.0 (0.893) NA 
Patient 4 46, XY 1.73 2,282.0 ND ND 1,578.0 (0.980) 491.0 (0.793) NA 
Patient 5 46, XY, t(3;5) 0.91 2,191.0 ND ND 578.0 (0.812) ND NA 
Patient 6 46, XY, +9 1.04 440.3 ND ND 175.2 (0.879) ND NA 
Patient 7 46, XY, t(8;21) 0.74 902.4 ND ND 138.5 (0.825) ND NA 
Patient 8 46, XX, t(8;21) 0.72 2,228.0 306.1 (0.346) 89.51 (0.459) 34.73 (0.714) ND NA 
Patient 9 46, XX, t(8;21) 0.18 495.9 9.177 (0.861) ND ND ND NA 

NOTE: Cytarabine IC50s are presented as mean plus SE from at least three independent experiments with the cell lines. Numbers in parentheses represent the CI values.

Abbreviations: NA, not applicable; ND, not determined.

To determine whether the synergistic antileukemic activity of VPA and cytarabine was unique to the THP-1 subline, analogous cytotoxicity experiments were done with the Kasumi-1, MV4-11, and CMS sublines derived from children with different AML subtypes. VPA showed variable cytotoxicities in the three additional AML sublines, with IC50s ranging from 0.37 to 2.7 mmol/L (Table 1). It is interesting that MV4-11 [harbors t(4;11)] and Kasumi-1 [harbors t(8;21)] cells were both substantially more sensitive to VPA than were the THP-1 and CMS sublines (Table 1). At 0.3 mmol/L VPA, simultaneous treatment with cytarabine resulted in 8.4- and 34.3-fold decreases in cytarabine IC50s, respectively, in Kasumi-1 and MV4-11 cells compared with those from cytarabine alone (Table 1). The results with the MV4-11 cells are particularly interesting because they harbor a FLT3 ITD in addition to t(4;11) (31). For CMS cells, simultaneous administration of VPA and cytarabine also resulted in a 2-fold decreased cytarabine IC50 at 1 mmol/L VPA compared with that from cytarabine alone (Table 1).

Analogous results were obtained when AML blasts collected at diagnosis from nine children with de novo AML were evaluated following cotreatment with cytarabine and VPA (0.15-1 mmol/L; Table 1). As with Kasumi-1 cells, diagnostic blasts from t(8;21) AML cases (n = 3, patients 7-9) were significantly more sensitive to VPA than non-t(8;21) AML blasts (n = 6, patients 1-6; median VPA IC50, 0.38 versus 1.41 mmol/L; P = 0.024; Table 1; Fig. 1E) and showed 6.5- to 64.1-fold decreased cytarabine IC50s when combined with VPA at doses 0.5 mmol/L or lower compared with that from cytarabine alone. By contrast, non-t(8;21) AML blasts only showed 1.3- to 13-fold decreases in cytarabine IC50s when combined with 0.5 mmol/L VPA (P = 0.024; Table 2; Fig. 1F).

For both AML cell lines and diagnostic blast samples, cytarabine and VPA were again synergistic by isobologram analyses (data not shown) and by CI values (Table 1). Collectively, our results show that synergistic antileukemic effects of combined cytarabine and VPA are broad ranging and occur in multiple AML subtypes.

VPA and cytarabine synergistically induce apoptosis of pediatric AML cells

We hypothesized that VPA may lower the apoptotic threshold in pediatric AML cells, rendering them more susceptible to apoptosis induced by cytarabine. Another possibility could be that VPA combines with cytarabine to induce cell cycle arrest, resulting in synergistic antileukemic activity on this basis. To test these hypotheses, THP-1 and Kasumi-1 cells, treated with cytarabine and VPA individually or in combination for 96 hours, were analyzed by flow cytometry to determine effects on cell cycle distribution and apoptosis. Treatment with cytarabine alone substantially induced apoptosis in both THP-1 and Kasumi-1 cells, whereas treatment with VPA by itself resulted in only marginally increased apoptosis in both cell lines (Fig. 2A and B). Combined VPA and cytarabine caused a substantial and synergistic induction of apoptosis compared with that resulting from the individual drug treatments (cooperativity index = 0.46 and 0.55, respectively; Fig. 2A and B).

Fig. 2.

VPA augments apoptosis and S-phase arrest induced by cytarabine in pediatric AML cells. A, B, and E, THP-1 cells (A), Kasumi-1 cells (B), and t(8;21) AML diagnostic blasts (E) were treated with cytarabine or VPA alone or in combination for 96, 96, and 24 h, respectively. Early and late apoptosis events in the cells were determined by Annexin V/PI staining and flow cytometry analyses. Data are presented as net percent of Annexin V+ cells relative to that of untreated cells. C and D, THP-1 (C) and Kasumi-1 cells (D) were treated with cytarabine or VPA alone or combined for 96 h. Cell cycle distribution was determined by PI staining and flow cytometry analysis.

Fig. 2.

VPA augments apoptosis and S-phase arrest induced by cytarabine in pediatric AML cells. A, B, and E, THP-1 cells (A), Kasumi-1 cells (B), and t(8;21) AML diagnostic blasts (E) were treated with cytarabine or VPA alone or in combination for 96, 96, and 24 h, respectively. Early and late apoptosis events in the cells were determined by Annexin V/PI staining and flow cytometry analyses. Data are presented as net percent of Annexin V+ cells relative to that of untreated cells. C and D, THP-1 (C) and Kasumi-1 cells (D) were treated with cytarabine or VPA alone or combined for 96 h. Cell cycle distribution was determined by PI staining and flow cytometry analysis.

Close modal

As expected, treatment of THP-1 and Kasumi-1 cells with cytarabine alone resulted in S- and G2-M–phase blockade compared with untreated cells (Fig. 2C and D). Treatment with VPA by itself caused arrest in G1-S progression in THP-1 cells (Fig. 2C). However, VPA treatment of Kasumi-1 cells caused at most marginal effects on cell cycle progression (e.g., slight increase of G1 phase and slight decrease of S phase; Fig. 2D). In both cell lines, cotreatment with VPA and cytarabine resulted in additional S arrest compared with that from cytarabine alone; in THP-1 cells, combined treatment resulted in an abrogation of the G1 arrest by VPA alone (Fig. 2C and D). These results show that VPA augments both apoptosis and S-phase arrest induced by cytarabine in THP-1 and Kasumi-1 cells.

To extend these latter results to diagnostic AML patient samples, blasts from patient 7 (Table 1) for which there were sufficient cells were treated with cytarabine and VPA individually or in combination for 24 hours and analyzed by flow cytometry for apoptosis and cell cycle distribution. Again, there was a synergistic induction of apoptosis by combined cytarabine and VPA (cooperativity index = 0.82; Fig. 2E). Changes in cell cycle distribution in the blasts could not be determined due to lack of cell proliferation (data not shown).

Cytarabine and VPA synergistically activate caspase-9 and caspase-3 in pediatric AML cells

To determine if apoptosis induced by cytarabine and VPA was associated with caspase activation, THP-1 and Kasumi-1 cells treated with cytarabine and VPA alone or combined for 96 hours were subjected to caspase-9 and caspase-3 enzymatic assays. In Fig. 3, cotreatments with cytarabine and VPA resulted in synergistic activation of caspase-9 and caspase-3 in both cell lines. These results show that cytarabine and VPA synergistically induce apoptosis of pediatric AML cells through the intrinsic apoptotic pathway.

Fig. 3.

Synergistic activation of caspase-9 and caspase-3 by cytarabine and VPA in THP-1 and Kasumi-1 cells. Whole-cell lysates from Kasumi-1 (A and B) and THP-1 (C and D) cells treated with cytarabine or VPA alone or in combination for 96 h were subjected to caspase-9 and caspase-3 enzymatic assays, respectively, as described in Materials and Methods. THP-1 and Kasumi-1 cells treated with 500 and 1,000 nmol/L daunorubicin, respectively, for 16 h were used as the positive controls.

Fig. 3.

Synergistic activation of caspase-9 and caspase-3 by cytarabine and VPA in THP-1 and Kasumi-1 cells. Whole-cell lysates from Kasumi-1 (A and B) and THP-1 (C and D) cells treated with cytarabine or VPA alone or in combination for 96 h were subjected to caspase-9 and caspase-3 enzymatic assays, respectively, as described in Materials and Methods. THP-1 and Kasumi-1 cells treated with 500 and 1,000 nmol/L daunorubicin, respectively, for 16 h were used as the positive controls.

Close modal

VPA and cytarabine cooperatively induce DNA damage in THP-1 and Kasumi-1 cells

Efforts were then undertaken to determine the molecular mechanisms that underlie the synergistic induction of apoptosis by the two agents. Cytarabine is a DNA-damaging agent that causes DNA double-strand breaks. A previous study suggested that HDACIs can also cause DNA damage in leukemia cells (9). Thus, we hypothesized that cytarabine and VPA cooperate in causing DNA damage, which subsequently triggers apoptosis. To test this possibility, THP-1 and Kasumi-1 cells were treated with variable concentrations of cytarabine or VPA, alone or combined, for 96 hours, and protein lysates were subjected to Western blotting to detect γH2AX, a biomarker of DNA double-strand breaks (32). Interestingly, cotreatment with VPA and cytarabine resulted in distinctly cooperative induction of γH2AX in both cell lines (Fig. 4A). In Kasumi-1 cells, this cooperative induction of γH2AX was both cytarabine and VPA concentration dependent (Fig. 4B). These results establish that VPA augments cytarabine-induced DNA double-strand breaks, which may trigger apoptosis. It is important to note that there was no difference in the extent of synergy of VPA (0.5 mmol/L) with 100 or 200 nmol/L cytarabine in terms of triggering DNA damage. This suggests that combining the two agents would allow for a dose reduction in cytarabine.

Fig. 4.

Cooperative induction of DNA double-strand breaks by VPA and cytarabine in THP-1 and Kasumi-1 cells. A, whole-cell lysates were prepared from Kasumi-1 (top) and THP-1 (bottom) cells treated with VPA and cytarabine alone or in combination for 96 h and subjected to Western blotting probed by anti-γH2AX or anti-actin antibody. B, Kasumi-1 cells were treated with variable concentrations of cytarabine and fixed concentration of VPA or variable concentrations of VPA and fixed concentration of cytarabine alone or in combination for 96 h. Whole-cell lysates were extracted and subjected to Western blotting probed by anti-γH2AX or anti-actin antibody. C and D, Kasumi-1 cells were treated with combined cytarabine and VPA for up to 96 h and cell lysates were extracted and subjected to Western blotting probed by anti-γH2AX or anti-actin antibody (C) or to caspase-3 assays as described in Materials and Methods (D). E and F, the relationships between the levels for γH2AX and the activities of caspase-3 in Kasumi-1 cells treated with combined cytarabine and VPA for up to 48 h (E) or 96 h (F) were determined by the Pearson tests.

Fig. 4.

Cooperative induction of DNA double-strand breaks by VPA and cytarabine in THP-1 and Kasumi-1 cells. A, whole-cell lysates were prepared from Kasumi-1 (top) and THP-1 (bottom) cells treated with VPA and cytarabine alone or in combination for 96 h and subjected to Western blotting probed by anti-γH2AX or anti-actin antibody. B, Kasumi-1 cells were treated with variable concentrations of cytarabine and fixed concentration of VPA or variable concentrations of VPA and fixed concentration of cytarabine alone or in combination for 96 h. Whole-cell lysates were extracted and subjected to Western blotting probed by anti-γH2AX or anti-actin antibody. C and D, Kasumi-1 cells were treated with combined cytarabine and VPA for up to 96 h and cell lysates were extracted and subjected to Western blotting probed by anti-γH2AX or anti-actin antibody (C) or to caspase-3 assays as described in Materials and Methods (D). E and F, the relationships between the levels for γH2AX and the activities of caspase-3 in Kasumi-1 cells treated with combined cytarabine and VPA for up to 48 h (E) or 96 h (F) were determined by the Pearson tests.

Close modal

Induction of γH2AX by combined VPA/cytarabine was an early molecular event in Kasumi-1 cells, as revealed by a time course study (Fig. 4C). Thus, substantial induction of γH2AX (2.6-fold increase relative to control) was detected by Western blotting as early as at 1.5 hours (Fig. 4C), accompanied by caspase-3 activation starting at 6 hours (Fig. 4D). Further, the levels of γH2AX significantly correlated with caspase-3 activities over 48 hours (r = 0.90, P = 006; Fig. 4E). However, this association was abolished when the 96-hour time data were included (r = 0.68, P = 0.06; Fig. 4F). These results strongly suggest that DNA damage was associated with caspase-3 activation in Kasumi-1 cells treated with combined cytarabine and VPA during early times (within 48 hours). There may be other factor(s) contributing to the late time (96 hours) caspase-3 activation in this experiment.

Bim is a critical determinant of apoptosis induced by cytarabine and combined VPA and cytarabine in pediatric AML cells

Previous studies showed that HDACIs can induce Bim to promote apoptosis in cancer cells (33, 34). It is conceivable that VPA also induces Bim expression in pediatric AML cells, thus contributing to apoptosis induced by combined VPA and cytarabine. As shown in Fig. 5A and B, modest induction of the BimEL isoform by VPA and VPA plus cytarabine was detected in both THP-1 and Kasumi-1 cells. In contrast, levels of other Bcl-2 family proteins were largely unchanged (Supplementary Fig. S1). These results suggest that Bim could be another important determinant of the antileukemic activities of combined VPA/cytarabine in pediatric AML cells. In contrast to the DNA damage response, induction of Bim seemed to be a later molecular event in both sublines (after 48 hours of treatment; Fig. 5C). This could explain the disproportionately increased caspase-3 activation seen at later times in Kasumi-1 cells (48 and 96 hours; Fig. 4D).

Fig. 5.

Bim plays a critical role in apoptosis induced by cytarabine and cytarabine plus VPA in pediatric AML cells. A and B, Kasumi-1 (A) and THP-1 (B) cells were treated with cytarabine or VPA alone or in combination for 96 h. Whole-cell lysates were extracted and subjected to Western blotting probed by anti-Bim or anti-actin antibody. C, Kasumi-1 and THP-1 cells were treated with combined cytarabine and VPA for up to 96 h and cell lysates were extracted and subjected to Western blotting probed by anti-Bim or anti-actin antibody. D and E, THP-1 cells were infected by Bim or negative (NTC) control shRNA lentivirus clones. After selection with puromycin, infected THP-1 cells were expanded and treated with cytarabine or VPA alone or combined for 96 h. The treated cells were then subjected to Western blotting for Bim expression (C) and to flow cytometry analysis for apoptosis (D).

Fig. 5.

Bim plays a critical role in apoptosis induced by cytarabine and cytarabine plus VPA in pediatric AML cells. A and B, Kasumi-1 (A) and THP-1 (B) cells were treated with cytarabine or VPA alone or in combination for 96 h. Whole-cell lysates were extracted and subjected to Western blotting probed by anti-Bim or anti-actin antibody. C, Kasumi-1 and THP-1 cells were treated with combined cytarabine and VPA for up to 96 h and cell lysates were extracted and subjected to Western blotting probed by anti-Bim or anti-actin antibody. D and E, THP-1 cells were infected by Bim or negative (NTC) control shRNA lentivirus clones. After selection with puromycin, infected THP-1 cells were expanded and treated with cytarabine or VPA alone or combined for 96 h. The treated cells were then subjected to Western blotting for Bim expression (C) and to flow cytometry analysis for apoptosis (D).

Close modal

To provide direct evidence that Bim is a critical effector of the antileukemic activities of cytarabine with and without VPA, lentivirus shRNA knockdown of Bim was done in THP-1 cells. shRNA knockdown of Bim (∼40%) substantially abolished its induction by VPA and combined VPA/cytarabine (Fig. 5D). This was accompanied by significantly decreased apoptosis induced by cytarabine alone and combined cytarabine/VPA (Fig. 5E).

Collectively, these results strongly support our hypothesis that cytarabine and VPA cause DNA double-strand breaks in a cooperative fashion, which in turn triggers caspase activation and apoptosis. Further, VPA induces expression of Bim, which promotes apoptosis induced by cytarabine.

HDAC inhibition represents one of the most promising epigenetic treatments for cancer because HDACIs have been established to reactivate silenced genes and exert pleiotropic antitumor effects selectively in cancer cells (5). The ability of HDACIs to induce cell differentiation, cell cycle arrest, and apoptosis in human leukemic cells but not in normal cells has stimulated significant interest in clinical applications as antileukemic agents (5, 1820). Currently, HDACIs including the antiepileptic agent VPA are being evaluated in the treatment of acute leukemias (1315). Despite their well-characterized molecular and cellular effects, single-agent activity of this class of drugs has been modest (5). Accordingly, there has been significant interest in developing rationally designed combination therapies using HDACIs.

In this study, we analyzed the cellular and molecular effects of combined cytarabine and VPA in a panel of clinically relevant pediatric AML cell lines and diagnostic blasts from children with de novo AML. Our rationale was based on the central role of cytarabine in AML chemotherapy (13) and on the documented ability of VPA to induce apoptosis specifically in leukemia cells, without causing proliferation inhibition of normal hematopoietic progenitor cells (35). Indeed, phase I/II studies using VPA as a single agent for adults with refractory AML or myelodysplastic syndrome have shown that VPA is well tolerated (15, 36).

The activity of VPA alone or in combination with cytarabine was initially evaluated against THP-1 AML cells, the most cytarabine-resistant subline tested in our study. In vitro incubations of THP-1 cells with VPA resulted in inhibition of cell proliferation in a dose-dependent manner, accompanied by hyperacetylation of histones H3 and H4. Interestingly, when VPA was incubated simultaneously with cytarabine, there was a synergistic loss of cell proliferation. When this was expanded to include three additional cell lines derived from children with different AML subtypes, synergism was again shown, suggesting that this mechanism may be broadly applicable to pediatric AMLs. Further, synergistic interactions between VPA and cytarabine were observed in nine diagnostic blast samples from children with AML. Of particular interest, t(8;21) AML cells were significantly more sensitive to VPA and showed the greatest response to cotreatment with cytarabine and VPA. This was not unexpected, given that several fusion proteins (AML-1/ETO, PML-RARA, etc.) recruit nuclear corepressor complexes (which contain HDACs; ref. 7). Thus, AML cases harboring these fusion genes might be preferentially susceptible to HDACIs. Previous pharmacokinetic studies have shown that clinically achievable trough levels of VPA used in the treatment of children with epilepsy (37) approximate the in vitro concentrations of VPA that synergized with cytarabine in our study.

The synergistic cytotoxicity of combined cytarabine and VPA is clearly due to cell death because synergistic induction of apoptosis by the two agents in both pediatric AML cell lines and diagnostic blasts was detected. In THP-1 cells, VPA inhibited cell cycle progression at G1-S, which may block apoptosis mediated by the HDACI (38). Interestingly, combined cytarabine and VPA completely abolished VPA-induced G1 arrest and resulted in additional S-phase arrest, which may favor apoptosis induced by cotreatment with these agents.

Our mechanistic studies in THP-1 and Kasumi-1 cells suggested that induction of apoptosis through caspase activation directly contributed to the potent synergism between cytarabine and VPA. Interestingly, this was accompanied by cooperative induction of DNA double-strand breaks, as reflected by the induction of γH2AX. Induction of γH2AX was significantly associated with caspase-3 activation, suggesting that DNA double-strand breaks were responsible for the apoptotic response on treatment with the two agents. However, the molecular mechanism(s) underlying VPA-induced DNA damage in pediatric AML cells remains elusive. Additional studies are under way to further determine the effects of HDACIs in inducing DNA damage in this disease.

Besides induction of DNA damage, both VPA and combined VPA/cytarabine also induced expression of the BH3-only proapoptotic protein Bim in both Kasumi-1 and THP-1 cells. Bim has been classified as an “activator” in view of its purported ability to engage directly and activate Bax and Bak (39). It has been well documented that Bim is critical for HDACI-induced apoptosis of both solid tumor and leukemia cells (33, 34). In this study, we showed that Bim also plays critical roles in cytarabine-induced and cytarabine plus VPA–induced apoptosis in pediatric AML cells. However, Bim may not be responsible for the synergy between the two agents because only VPA, but not cytarabine, induced Bim expression in our experiments.

Together, our results document global synergistic antileukemic activities of combined VPA/cytarabine in pediatric AMLs and suggest that VPA could be an attractive agent for combination therapy of this deadly disease. Based on our results, VPA was recently incorporated into one of the treatment arms for high-risk AML in the St. Jude Children's Research Hospital AML08 clinical trial “A Randomized Trial of Clofarabine Plus Cytarabine Versus Conventional Induction Therapy and of Natural Killer Cell Transplantation Versus Conventional Consolidation Therapy in Patients with Newly Diagnosed Acute Myeloid Leukemia.” In this trial, children with acute megakaryocytic leukemia without t(1;22) and other high-risk patients without FLT3-ITD will receive a combination of VPA with low-dose cytarabine, daunorubicin, and etoposide (LD-ADE) during the second induction course. The incorporation of VPA as a new agent for treating high-risk AML patients has potential advantages based on its well-characterized toxicity profile and safety in children. Based on our results, incorporation of VPA into cytarabine-based clinical trials for treatment of different risk groups of pediatric AML should be strongly considered.

No potential conflicts of interest were disclosed.

Grant Support: Karmanos Cancer Institute Start-up Fund, Children's Research Center of Michigan, Leukemia Research Life, Herrick Foundation, Children's Leukemia Foundation of Michigan, National Cancer Institute grant CA120772, Leukemia and Lymphoma Society, ELANA Fund, Justin's Gift Charity, Sehn Family Foundation, St. Baldrick's Foundation, Dale Meyer Memorial Endowment for Leukemia Research, Ring Screw Textron Endowed Chair for Pediatric Cancer Research (J.W. Taub), and Natural Science Foundation of China grant NSFC30873093.

The costs of publication of this article were defrayed in part by the payment of page charges. This article must therefore be hereby marked advertisement in accordance with 18 U.S.C. Section 1734 solely to indicate this fact.

1
Meshinchi
S
,
Arceci
RJ
. 
Prognostic factors and risk-based therapy in pediatric acute myeloid leukemia
.
Oncologist
2007
;
12
:
341
55
.
2
Kaspers
GJ
,
Zwaan
CM
. 
Pediatric acute myeloid leukemia: towards high-quality cure of all patients
.
Haematologica
2007
;
92
:
1519
32
.
3
Zwaan
CM
,
Kaspers
GJ
. 
Possibilities for tailored and targeted therapy in paediatric acute myeloid leukaemia
.
Br J Haematol
2004
;
127
:
264
79
.
4
Grant
S
. 
Ara-C: cellular and molecular pharmacology
.
Adv Cancer Res
1998
;
72
:
197
233
.
5
Bolden
JE
,
Peart
MJ
,
Johnstone
RW
. 
Anticancer activities of histone deacetylase inhibitors
.
Nat Rev Drug Discov
2006
;
5
:
769
84
.
6
Wang
ZY
,
Chen
Z
. 
Acute promyelocytic leukemia: from highly fatal to highly curable
.
Blood
2008
;
111
:
2505
15
.
7
Berman
JN
,
Look
AT
. 
Targeting transcription factors in acute leukemia in children
.
Curr Drug Targets
2007
;
8
:
727
37
.
8
Gao
N
,
Rahmani
M
,
Shi
X
,
Dent
P
,
Grant
S
. 
Synergistic antileukemic interactions between 2-medroxyestradiol (2-ME) and histone deacetylase inhibitors involve Akt down-regulation and oxidative stress
.
Blood
2006
;
107
:
241
9
.
9
Gaymes
TJ
,
Padua
RA
,
Pla
M
, et al
. 
Histone deacetylase inhibitors (HDI) cause DNA damage in leukemia cells: a mechanism for leukemia-specific HDI-dependent apoptosis?
Mol Cancer Res
2006
;
4
:
563
73
.
10
Ellis
L
,
Pan
Y
,
Smyth
GK
, et al
. 
Histone deacetylase inhibitor panobinostat induces clinical responses with associated alterations in gene expression profiles in cutaneous T-cell lymphoma
.
Clin Cancer Res
2008
;
14
:
4500
10
.
11
Kelly
WK
,
O'Connor
OA
,
Krug
LM
, et al
. 
Phase I study of an oral histone deacetylase inhibitor, suberoylanilide hydroxamic acid, in patients with advanced cancer
.
J Clin Oncol
2005
;
23
:
3923
31
.
12
Kuendgen
A
,
Strupp
C
,
Aivado
M
, et al
. 
Treatment of myelodysplastic syndromes with valproic acid alone or in combination with all-trans retinoic acid
.
Blood
2004
;
104
:
1266
9
.
13
Cimino
G
,
Lo-Coco
F
,
Fenu
S
, et al
. 
Sequential valproic acid/all-trans retinoic acid treatment reprograms differentiation in refractory and high-risk acute myeloid leukemia
.
Cancer Res
2006
;
66
:
8903
11
.
14
Garcia-Manero
G
,
Kantarjian
HM
,
Sanchez-Gonzalez
B
, et al
. 
Phase 1/2 study of the combination of 5-aza-2′-deoxycytidine with valproic acid in patients with leukemia
.
Blood
2006
;
108
:
3271
9
.
15
Soriano
AO
,
Yang
H
,
Faderl
S
, et al
. 
Safety and clinical activity of the combination of 5-azacytidine, valproic acid, and all-trans retinoic acid in acute myeloid leukemia and myelodysplastic syndrome
.
Blood
2007
;
110
:
2302
8
.
16
Munster
P
,
Marchion
D
,
Bicaku
E
, et al
. 
Phase I trial of histone deacetylase inhibition by valproic acid followed by the topoisomerase II inhibitor epirubicin in advanced solid tumors: a clinical and translational study
.
J Clin Oncol
2007
;
25
:
1979
85
.
17
Gottlicher
M
,
Minucci
S
,
Zhu
P
, et al
. 
Valproic acid defines a novel class of HDAC inhibitors inducing differentiation of transformed cells
.
EMBO J
2001
;
20
:
6969
78
.
18
Duenas-Gonzalez
A
,
Candelaria
M
,
Perez-Plascencia
C
,
Perez-Cardenas
E
,
de la Cruz-Hernandez
E
,
Herrera
LA
. 
Valproic acid as epigenetic cancer drug: preclinical, clinical and transcriptional effects on solid tumors
.
Cancer Treat Rev
2008
;
34
:
206
22
.
19
Tang
R
,
Faussat
AM
,
Majdak
P
, et al
. 
Valproic acid inhibits proliferation and induces apoptosis in acute myeloid leukemia cells expressing P-gp and MRP1
.
Leukemia
2004
;
18
:
1246
51
.
20
Insinga
A
,
Monestiroli
S
,
Ronzoni
S
, et al
. 
Inhibitors of histone deacetylases induce tumor-selective apoptosis through activation of the death receptor pathway
.
Nat Med
2005
;
11
:
71
6
.
21
Sanchez-Gonzalez
B
,
Yang
H
,
Bueso-Ramos
C
, et al
. 
Antileukemia activity of the combination of an anthracycline with a histone deacetylase inhibitor
.
Blood
2006
;
108
:
1174
82
.
22
Yang
H
,
Hoshino
K
,
Sanchez-Gonzalez
B
,
Kantarjian
H
,
Garcia-Manero
G
. 
Antileukemia activity of the combination of 5-aza-2′-deoxycytidine with valproic acid
.
Leuk Res
2005
;
29
:
739
48
.
23
ten Cate
B
,
Samplonius
DF
,
Bijma
T
,
de Leij
LF
,
Helfrich
W
,
Bremer
E
. 
The histone deacetylase inhibitor valproic acid potently augments gemtuzumab ozogamicin-induced apoptosis in acute myeloid leukemic cells
.
Leukemia
2007
;
21
:
248
52
.
24
Miller
CP
,
Ban
K
,
Dujka
ME
, et al
. 
NPI-0052, a novel proteasome inhibitor, induces caspase-8 and ROS-dependent apoptosis alone and in combination with HDAC inhibitors in leukemia cells
.
Blood
2007
;
110
:
267
77
.
25
Siitonen
T
,
Koistinen
P
,
Savolainen
ER
. 
Increase in Ara-C cytotoxicity in the presence of valproate, a histone deacetylase inhibitor, is associated with the concurrent expression of cyclin D1 and p27(Kip 1) in acute myeloblastic leukemia cells
.
Leuk Res
2005
;
29
:
1335
42
.
26
Taub
JW
,
Huang
X
,
Matherly
LH
, et al
. 
Expression of chromosome 21-localized genes in acute myeloid leukemia: differences between Down syndrome and non-Down syndrome blast cells and relationship to in vitro sensitivity to cytosine arabinoside and daunorubicin
.
Blood
1999
;
94
:
1393
400
.
27
Tallarida
RJ
. 
Drug synergism: its detection and applications
.
J Pharmacol Exp Ther
2001
;
298
:
865
72
.
28
Chou
TC
. 
Theoretical basis, experimental design, and computerized simulation of synergism and antagonism in drug combination studies
.
Pharmacol Rev
2006
;
58
:
621
81
.
29
Edwards
H
,
Xie
C
,
LaFiura
KM
, et al
. 
RUNX1 regulates phosphoinositide 3-kinase/AKT pathway: role in chemotherapy sensitivity in acute megakaryocytic leukemia
.
Blood
2009
;
114
:
2744
52
.
30
Ge
Y
,
Stout
ML
,
Tatman
DA
, et al
. 
GATA1, cytidine deaminase, and the high cure rate of Down syndrome children with acute megakaryocytic leukemia
.
J Natl Cancer Inst
2005
;
97
:
226
31
.
31
Quentmeier
H
,
Reinhardt
J
,
Zaborski
M
,
Drexler
HG
. 
FLT3 mutations in acute myeloid leukemia cell lines
.
Leukemia
2003
;
17
:
120
4
.
32
Paull
TT
,
Rogakou
EP
,
Yamazaki
V
,
Kirchgessner
CU
,
Gellert
M
,
Bonner
WM
. 
A critical role for histone H2AX in recruitment of repair factors to nuclear foci after DNA damage
.
Curr Biol
2000
;
10
:
886
95
.
33
Chen
S
,
Dai
Y
,
Pei
XY
,
Grant
S
. 
Bim upregulation by histone deacetylase inhibitors mediates interactions with the Bcl-2 antagonist ABT-737: evidence for distinct roles for Bcl-2, Bcl-xL, Mcl-1
.
Mol Cell Biol
2009
;
29
:
6149
69
.
34
Zhao
Y
,
Tan
J
,
Zhuang
L
,
Jiang
X
,
Liu
ET
,
Yu
Q
. 
Inhibitors of histone deacetylases target the Rb-E2F1 pathway for apoptosis induction through activation of proapoptotic protein Bim
.
Proc Natl Acad Sci U S A
2005
;
102
:
16090
5
.
35
Kawagoe
R
,
Kawagoe
H
,
Sano
K
. 
Valproic acid induces apoptosis in human leukemia cells by stimulating both caspase-dependent and -independent apoptotic signaling pathways
.
Leuk Res
2002
;
26
:
495
502
.
36
Kuendgen
A
,
Schmid
M
,
Schlenk
R
, et al
. 
The histone deacetylase (HDAC) inhibitor valproic acid as monotherapy or in combination with all-trans retinoic acid in patients with acute myeloid leukemia
.
Cancer
2006
;
106
:
112
9
.
37
Gerstner
T
,
Bell
N
,
Longin
E
,
Konig
SA
. 
Oral rapid loading of valproic acid—an alternative to the usual saturation scheme?
Seizure
2006
;
15
:
630
2
.
38
Weiss
RH
. 
p21Waf1/Cip1 as a therapeutic target in breast and other cancers
.
Cancer Cell
2003
;
4
:
425
9
.
39
Letai
A
. 
BCL-2: found bound and drugged!
Trends Mol Med
2005
;
11
:
442
4
.