ARTICLE

Selective Efficacy of Depsipeptide in a Xenograft Model of Epstein-Barr Virus–Positive Lymphoproliferative Disorder

Sameek Roychowdhury, Robert A. Baiocchi, Srinivas Vourganti, Darshna Bhatt, Bradley W. Blaser, Aharon G. Freud, Jason Chou, Chang-Shi Chen, Jim J. Xiao, Mark Parthun, Kenneth K. Chan, Charles F. Eisenbeis, Amy K. Ferketich, Michael R. Grever, Ching-Shih Chen, Michael A. Caligiuri

Affiliations of authors: Department of Molecular Virology, Immunology, and Medical Genetics (SR, MAC), Medical Scientist Program (SR, BWB, AGF, MAC), Department of Internal Medicine (RAB, SV, DB, JC, KKC, CFE, MRG, MAC), Division of Hematology/Oncology (RAB, JC, CFE, MRG, MAC), Integrated Biomedical Graduate Program (BWB, AGF), College of Pharmacy (Chang-Shi Chen, JJX, KKC, Ching-Shih Chen), Comprehensive Cancer Center (JJX, KKC, CFE, Ching-Shih Chen, MAC), Experimental Therapeutics Program (KKC, MP, MRG), Department of Molecular and Cellular Biochemistry (MP), Division of Epidemiology and Biometrics (AKF), The Ohio State University, Columbus

Correspondence to: Michael A. Caligiuri, MD, A458 Starling Loving Hall, 320 W. 10th Ave., Columbus, OH 43210 (e-mail: caligiuri-1{at}medctr.osu.edu)


    ABSTRACT
 Top
 Notes
 Abstract
 Introduction
 Materials and Methods
 Results
 Discussion
 References
 
Background: Immune-compromised individuals are at increased risk for developing aggressive Epstein-Barr virus (EBV)–associated lymphoproliferative disorders after primary EBV infection or for reactivation of a preexisting latent EBV infection. We evaluated the effect of depsipeptide, a histone deacetylase inhibitor, on EBV-positive lymphoblastoid cell lines (LCLs) and Burkitt lymphoma cell lines in a mouse model and explored its mechanism of action in vitro. Methods: We studied EBV-transformed LCLs, which express a latent III (Lat-III) viral gene profile, as do some EBV-positive lymphoproliferative malignancies, and Burkitt lymphoma cell lines, which express a Lat-I viral gene profile. Cell lines were used to characterize depsipeptide-induced apoptosis, which was evaluated by flow cytometry. Flow cytometry, western blot analyses, and histone deacetylase inhibitors were used to investigate components of prodeath and survival pathways in vitro. We studied depsipeptide’s effects on survival with a mouse xenograft model of EBV-positive human B-cell tumors (groups of 10 mice). All statistical tests were two-sided. Results: Depsipeptide (5 mg/m2 of body surface area) treatment was associated with statistically significantly improved survival of mice carrying Lat-III EBV–positive LCL tumors, compared with that of control-treated mice (day 30: for depsipeptide-treated mice, 90% survival, 95% confidence interval [CI] = 73.2% to 100%; for control-treated mice, 20% survival, 95% CI = 5.79% to 69.1%; P<.001), but it was not associated with survival of mice carrying Lat-I EBV–positive Burkitt lymphoma tumors. Depsipeptide induced apoptosis in 64% of LCLs and in 14% of EBV-positive Burkitt lymphoma cells in vitro. Depsipeptide-treated LCL cultures had two distinct cell populations—one sensitive and one resistant to depsipeptide. Depsipeptide-mediated apoptosis was associated with a 12-fold increased level of active caspase 3, but some apoptosis persisted despite z-VAD-fmk treatment to inhibit caspase activity. Depsipeptide-resistant LCLs expressed higher levels of latent membrane protein 1 (LMP1; P = .017), BCL2 (P = .032), and nuclear factor {kappa}B (NF-{kappa}B) (P<.001) than depsipeptide-sensitive LCLs; this resistance was circumvented by treatment with PS-1145, an inhibitor of NF-{kappa}B activation (P<.001). Conclusions: Apoptosis is induced by depsipeptide via caspase-dependent and -independent pathways in Lat-III EBV–positive LCLs and is enhanced by inhibiting NF-{kappa}B activity. Depsipeptide as a treatment for Lat-III EBV–associated lymphoproliferative disorders should be explored further in clinical trials.



    INTRODUCTION
 Top
 Notes
 Abstract
 Introduction
 Materials and Methods
 Results
 Discussion
 References
 
Epstein-Barr virus (EBV) has established latent infection in more than 90% of adults worldwide (1). Primary infection of healthy, immune-competent individuals by EBV is typically subclinical but is occasionally associated with the development of infectious mononucleosis. After primary infection, EBV establishes a persistent latent infection in memory B cells as the result of a highly regulated viral latency program and an efficient cell-mediated immune response to the virus. Three latent gene programs exist, each associated with a definitive pattern of viral gene expression. EBV-positive resting B cells display a type I latent (Lat-I) gene expression profile that is characterized by the expression of Epstein–Barr nuclear antigen 1 (EBNA1) and latent membrane protein (LMP) 2a. EBV-positive B lymphocytes display a type II latent (Lat-II) gene profile that is characterized by the expression of EBNA1, LMP1 and LMP2a. EBV-positive human B lymphoblastoid cell lines (LCLs), capable of proliferating indefinitely in vitro, display a type III latent (Lat-III) profile that is characterized by the expression of EBNA1, -2, -3a, -3b, -3c, and -4 and LMP1, -2a, and -2b (1).

Immune-compromised individuals are at increased risk for developing aggressive EBV-associated lymphoproliferative disorders after primary EBV infection or reactivation of a preexisting latent EBV infection. EBV-associated malignancies are characterized by specific patterns of latent EBV gene expression. Burkitt lymphomas display a Lat-I expression profile, Hodgkin disease and nasopharyngeal carcinoma express a Lat-II expression profile, and EBV-associated lymphoproliferative disorders in individuals with an immune deficiency generally display a Lat-III expression profile (1). Because most EBV-positive malignancies are associated with high mortality, new treatment strategies that specifically target EBV-transformed cells are needed (1,2).

In the past 10 years, a novel class of anticancer drugs that inhibit histone deacetylase enzymes has been developed. Histone deacetylases participate in the remodeling of chromatin structure by removing acetyl groups from lysine residues in the amino-terminal region of histones. Histone deacetylation stabilizes chromatin structure and represses transcription. Inhibition of histone deacetylases allows multisubunit complexes with histone acetyltransferase activity to covalently modify core histones that in turn alter chromatin conformation, making promoter regions more accessible to transcription factors and thus more permissive to transcriptional activation. Differentiation, apoptosis, and/or cell cycle arrest have been induced in several cancer cell lines in vitro by histone deacetylase inhibitors (3). Depsipeptide is a histone deacetylase inhibitor that has shown promising anticancer activity and been safely administered to patients with refractory neoplasms (4) and to patients with cutaneous T-cell lymphomas (5).

To our knowledge, the effect of histone deacetylase inhibition on the survival of a xenograft mouse model of human B-cell lymphoma has not been explored. The purpose of this study was to evaluate the effects of depsipeptide, a histone deacetylase inhibitor, on EBV-positive lymphoblastoid and Burkitt lymphoma cell lines in a xenograft mouse model and to explore its mechanism of action.


    MATERIALS AND METHODS
 Top
 Notes
 Abstract
 Introduction
 Materials and Methods
 Results
 Discussion
 References
 
Cell Lines, Cell Culture, Cell Viability, and Cell Cycle Synchronization

Human blood samples were procured under an Institutional Review Board–approved protocol at The Ohio State University Hospitals. We used three different human EBV-positive LCLs, two of which (PA-LCL and RM-LCL) had been developed in vitro from the peripheral blood of two patients diagnosed with a post-transplant lymphoproliferative disorder, and the third (C7M3) had been derived from a severe combined immune-deficient (SCID) mouse engrafted with human peripheral blood lymphocytes from a healthy EBV-positive donor (6). EBV-negative Burkitt lymphoma cell lines (BL-41 and Ramos) and EBV-positive Burkitt lymphoma cell lines (Raji, Daudi, and Jijoye) were obtained from the American Type Culture Collection (Manassas, VA). All cell lines were cultured in standard growth medium containing RPMI 1640 medium supplemented with antibiotics (penicillin at 100 U/mL and streptomycin at 100 µg/mL) and 10% fetal bovine serum (Invitrogen, Rockville, MD). All cell lines were mycoplasma free and were maintained at 37 °C in a humidified atmosphere of 5% CO2 and 95% air. Viability and cell counts were determined by trypan blue dye exclusion.

For cell cycle synchronization experiments, cells were synchronized in G1 phase by an overnight exposure (10 hours) to aphidicolin at 5 µg/mL (Sigma, St. Louis, MO). The cell cycle distribution was then confirmed via flow cytometry to assess the DNA content of cells after propidium iodide staining as described previously (11). After this treatment, 80% of cells were in G1 phase.

Treatment With Histone Deacetylase Inhibitors

Stock solutions of 3 mM trichostatin A or 3 M sodium butyrate (Sigma) were prepared in dimethyl sulfoxide (DMSO). Depsipeptide (5 µM) (Fujisawa Pharmaceutical) was dissolved in acetonitrile. Cells were incubated for 4 hours with vehicle control (DMSO or acetonitrile), trichostatin A (3, 3 x 101, or 3 x 102 µM), sodium butyrate (3 x 103, 3 x 104, or 3 x 105 µM), or depsipeptide (1 x 10–2, 5 x 10–2, 1 x 10–1, or 5 x 10–1 µM) for 4 hours. Cells were then washed twice with phosphate-buffered saline (PBS) and plated in standard growth medium (2 mL of RPMI 1640 medium with 10% fetal bovine serum and antibiotics per well, in triplicate wells of a 24-well plate; Falcon). For apoptosis and proliferation assays, 0.5 x 106 to 1.0 x 106 cells were harvested at 0, 24, 48, or 72 hours after exposure to histone deacetylase inhibitors or the vehicle control.

Animal Model

Six- to eight-week-old female SCID mice were purchased from Taconic Farms (Germantown, NY) and were maintained in pathogen-free, isolated cages. To improve tumor engraftment, all animals were depleted of murine natural killer cells with a single intraperitoneal injection of 0.2 mg of rat anti–mouse interleukin 2 receptor {beta} monoclonal antibodies (TM{beta}1) 2 days before engraftment with human tumor cell lines, as described previously (7). To ensure the consistency of cellular preparations for all in vivo experiments, we first cultured each cell line until a total of 10 x 109 cells were obtained. Frozen stocks (3 x 107 cells per vial) were then prepared in DMSO freezing medium (Sigma), frozen under controlled rate at –86 °C (using Control rate freezing container; Nalgene), and stored in a vapor-phase liquid nitrogen environment (–200 °C). Before in vivo passage, cryopreserved cells were thawed, cultured for 10 days, and then resuspended in PBS at room temperature for injection. We performed cell–dose titration trials to determine an optimal dose of cells that would lead to equivalent fatal tumor burdens in all mice (data not shown) and found that the optimal dose for both C7M3 EBV-positive LCLs and Raji EBV-positive Burkitt lymphoma tumor cells was 10 x 106 cells. Without intervention, mice carrying these tumor cells at the optimal dose have a mean survival time of 20 days.

For in vivo treatment, depsipeptide was suspended with sonication at 0.96 mg/mL in a solution of 10% HCO-60 and PBS at room temperature. HCO-60 is an emulsifying agent (Lipo Chemicals, Paterson, NJ) designed for slow drug release. One milliliter of this depsipeptide solution was diluted with 0.5 mL of 10% ethanol–40% propylene glycol in PBS and stored at 4 °C (final concentrations = depsipeptide at 0.64 mg/mL, 66% HCO-60, 3.3% ethanol, and 13.3% propylene glycol). Each day, a depsipeptide working stock was prepared by diluting 0.492 mL of the 0.64-mg/mL solution with 1.6 mL of sterile PBS (final depsipeptide concentration = 0.15 mg/mL).

For toxicity studies, mice were injected intraperitoneally each day with depsipeptide at 3, 5, or 10 mg/m2 of body surface area (n = 3 mice per group). For pharmacokinetic studies, plasma levels of depsipeptide after a single injection of drug (5 mg/m2, n = 3 mice per time point) were determined with a sensitive electrospray liquid chromatography–tandem mass spectroscopy assay, as previously described (4,8). For therapeutic studies, mice with tumors (groups of 10 mice) were injected intraperitoneally with 0.2 mL of depsipeptide (0.03 mg per mouse = 5 mg/m2) or with 0.2 mL of vehicle control on days 2, 4, 7, and 10 after tumor inoculation. Animals were monitored daily for signs of tumor burden, including weight loss, ruffled coat, and distended abdomen. When mice showed signs of tumor burden, they were killed with a lethal dose of anesthesia (ketamine–xylazine) followed by cervical dislocation. All animal research was reviewed and approved by the University Laboratory Animal Resources at The Ohio State University.

Flow Cytometric Analysis

After treatment with a histone deacetylase inhibitor, 0.5 x 106 to 1 x 106 cells (C7M3, PA, RM, Raji, Daudi, Jijoye, BL41, and Ramos), were harvested, washed, and assayed for apoptosis by dual staining with Annexin V–coupled fluorescein isothiocyanate (FITC) and propidium iodide (BD Pharmingen, San Diego, CA). Cells were washed in ice-cold PBS and resuspended in Annexin V binding buffer containing 5 µL of Annexin V–FITC and 1.5 µL of propidium iodide (50 µg/mL). After a 10-minute incubation at room temperature in the dark, samples were analyzed on a FACScalibur flow cytometer (Becton Dickinson, San Diego, CA). Results were interpreted as follows: Annexin V–positive and propidium iodide–negative events were considered apoptotic events, Annexin V–positive and propidium iodide–positive events were considered nonviable (apoptotic or necrotic), and Annexin V–negative and propidium iodide–negative events were considered viable (9).

Intracellular staining was performed with monoclonal antibodies specific for active caspase 3 (FITC-conjugated from BD Pharmingen), LMP1 (phycoerythrin-conjugated from BD Pharmingen), BCL2 (detected by secondary staining with allophycocyanin-conjugated goat anti–mouse monoclonal antibody; Molecular Probes, Eugene, OR), and isotype control monoclonal antibodies (BD Pharmingen). Intracellular staining was performed with an intracellular flow cytometry kit (BD Pharmingen) as described previously (10). In brief, cells were simultaneously fixed, permeabilized with the Cytofix/CytoPerm reagent, and then washed in Perm/Wash buffer. Nonspecific binding of monoclonal antibodies was blocked by preincubating samples with unconjugated mouse immunoglobulin G (Sigma). Flow cytometric data were analyzed with CellQuest (Becton-Dickinson) and graphed with WinMDI (Scripps Research Institute). Data are expressed as mean fluorescent intensity, representing populations gated on the specific parameters indicated. For determination of cell cycle and DNA fragmentation status, we measured DNA content with propidium iodide staining of permeabilized cells as described previously (11). Cells were stained with 20 µL of propidium iodide (50 µg/mL) and 5 µL of RNase (10 mg/mL). Cells were protected from light by wrapping tubes in foil and stored for 2 hours at 4 °C before sample collection during flow cytometry. Cell cycle data were analyzed with ModFit (Verity Software House, Topsham, ME).

Proliferation Assay With [3H]Thymidine Incorporation

After a 4-hour treatment with vehicle or 10, 50, 100, or 500 nM depsipeptide, cells in 100 µL of standard growth medium (RPMI 1640 medium supplemented with 10% fetal bovine serum, penicillin, and streptomycin) were cultured in a 96-well U-bottom plate at 25 000 cells per well (six wells per treatment) for 60 hours. Cells in each well were incubated with 1 µCi of [3H]thymidine (NEN Life Sciences, Boston, MA) for an additional 12 hours at 37 °C, 5% CO2 and 95% HEPA (0.3-µm pore size)–filtered air. Mitomycin C blocks DNA replication, resulting in G1/G0 arrest, and was used as a control for growth inhibition. Cells were harvested onto nitrocellulose filters, and [3H]thymidine incorporation was measured with a Beckman Coulter liquid scintillation counter. The proliferation index was expressed as the mean counts per minute in the six replicate wells (with 95% confidence intervals [CIs]).

Effect of Caspase Inhibition on Depsipeptide-Mediated Apoptosis

The broad-spectrum caspase inhibitor z-VAD-fmk (Enzyme Product Systems, Livermore, CA) was dissolved in DMSO for stock solution (15 mM) and further diluted in standard growth medium at concentrations used in experiments. Cell lines were incubated with DMSO (vehicle control, diluted in complete cell culture medium) or with 10–150 µM z-VAD-fmk for 1 hour at 37 °C. Cells were then treated for 4 hours with acetonitrile vehicle (diluted 1 : 10 [vol/vol] in standard culture medium), 100 nM or 500 nM depsipeptide, or the agonistic anti-Fas (CD95) monoclonal antibody 7c11, as described previously (12), at 1 µg/mL. Cells were washed twice with sterile PBS, plated in growth medium containing the DMSO control or z-VAD-fmk concentrations in triplicate wells in a 24-well plate, and harvested after 24, 48, or 72 hours of culture. DNA content was analyzed by flow cytometry and agarose gel electrophoresis as described previously (13,14).

Western Blot Analysis

Cleaved (active) caspase 3 was measured by western blot analysis as previously described (15). Treated cells were collected, washed with PBS, resuspended in sodium dodecyl sulfate (SDS) gel-loading buffer (100 mM Tris–HCl [pH 6.8], 4% [wt/vol] SDS, 0.2% bromophenol blue, 20% [vol/vol] glycerol, and 200 mM dithiothreitol), sonicated with an ultrasonic sonicator for 5 seconds, and boiled for 5 minutes. After brief centrifugation (10 000g for 2 minutes at 4 °C), equivalent amounts 10 µg of soluble protein, as determined by the Bradford method, were resolved by SDS–polyacrylamide gel electrophoresis in 10% minigels and transferred to nitrocellulose membranes with the use of a semidry transfer cell (Bio-Rad). The membranes were washed twice with Tris-buffered saline (TBS; 0.3% Tris, 0.8% NaCl, and 0.02% KCl) containing 0.05% Tween 20 (TBST) and then incubated with TBS containing 5% nonfat dry milk for 60 minutes to block nonspecific antibody binding. Each membrane was then incubated at 4 °C for 12 hours with a primary antibody specific for caspase 3 (Santa Cruz Biotechnology, Santa Cruz, CA), which was diluted 1 : 1000 in TBS containing 1% nonfat dry milk. The membranes were washed twice with TBST and then incubated at room temperature for 1 hour with a horseradish peroxidase–conjugated goat anti–rabbit immunoglobulin G diluted 1 : 5000 in TBS containing 1% nonfat dry milk. The membranes were washed twice with TBST, and bound antibody was visualized by enhanced chemiluminescence (ECL) using ECL western blotting detection reagents (Amersham Pharmacia Biotech, Little Chalfont, U.K.).

For evaluation of histone acetylation, we purified histone protein fractions from LCL and Burkitt lymphoma cell lines (C7M3, PA, RM-LCL, Raji, Daudi, Jijoye, and BL41), and western blot analysis was performed as previously described (16). In brief, duplicate purified histone protein preparations were separated by sodium dodecyl sulfate–polyacrylamide gel electrophoresis. One of the gels was stained with Coomassie blue to compare total protein levels, and the other gel was blotted onto a nitrocellulose membrane for western blot analysis. Western blots were probed with antibodies that recognize acetylated lysine residues at positions 5, 8, 12, and 16 within the amino-terminal tail of histone H4 (all monoclonal antibodies from Serotec, Raleigh, NC). Western blots were visualized by ECL as described in the manufacturer’s instructions (Amersham, Piscataway, NJ).

Nuclear Factor {kappa}B Detection and Inhibition

The p65 subunit of nuclear factor {kappa}B (NF-{kappa}B) was measured in the nuclear protein fraction of depsipeptide-treated cells and untreated control cells by use of an NF-{kappa}B Trans-Am kit (Active Motif, Carlsbad, CA). Cells were harvested after a 4-hour treatment with depsipeptide. For depsipeptide-treated EBV-positive LCLs (C7M3, PA, and RM), viable cells were isolated by centrifugation of suspended cells over a 5-mL Ficoll–Hypaque (Sigma) layer for 30 minutes (25 °C); cell viability was verified by trypan blue dye exclusion. Nuclear extracts were prepared from 5 x 106 to 10 x 106 cells with the NE-PER extraction kit, as described by the manufacturer (Pierce Biotechnology, Rockford, IL). Nuclear proteins (1 µg per well, measured with a standard Bradford protein assay) were loaded into quadruplicate wells of a Trans-Am 96-well plate and incubated for 1 hour at room temperature. These plates were coated by the manufacturer with NF-{kappa}B consensus oligonucleotides that serve as immobilized substrates to which active NF-{kappa}B complexes bind. Plates were washed and probed with anti–p65 subunit primary monoclonal antibodies and then with horseradish peroxidase–conjugated secondary monoclonal antibodies. After reactions with the horseradish peroxidase substrate and then with stopping reagents provided by the kit, absorbance at 450 nm was read with a spectrophotometer (Perkin-Elmer). Tumor necrosis factor {alpha}–stimulated HeLa cell extracts were used as positive controls, as described in the manufacturer’s recommendations. To determine specificity, we added wild-type and mutant NF-{kappa}B consensus oligonucleotides to HeLa extracts as additional negative and positive controls, respectively. Relative NF-{kappa}B activity was expressed as the average absorbance units per microgram of protein.

PS-1145 is a small molecule (PS-1145 dihydrochloride salt, molecular weight = 395.67; a gift from Millennium Pharmaceuticals) that specifically blocks NF-{kappa}B activation by inhibiting I{kappa}B kinase–mediated phosphorylation of I{kappa}B{alpha}, the key negative regulator of NF-{kappa}B activity (17). This inhibitor allows I{kappa}B{alpha} to evade proteasomal degradation and form stable heterodimers and/or homodimers with NF-{kappa}B subunits, preventing its translocation to the nucleus. Stock solutions of PS-1145 were made by dissolving lyophilized drug in DMSO to a final concentration of 10 mM. For experiments involving inhibition of NF-{kappa}B, cells were pretreated with 20 µM PS-1145 for 1 hour at 37 °C and then treated with depsipeptide or vehicle control for 4 hours at 37 °C. After treatment, cells were washed free of depsipeptide and PS-1145 twice with sterile PBS and then plated in growth medium containing 20 µM PS-1145 or control.

Statistical Analysis

The mean survival time, 95% confidence interval, and median survival time were calculated for the groups treated with depsipeptide and vehicle control by use of the Kaplan–Meier estimate (18). The survival times of the groups were compared with the log-rank test (18). This test was performed for each animal model (C7M3 and Raji). Data sets produced in vitro were analyzed with two-sample Student’s t tests with unequal variances. All statistical tests were two-sided.


    RESULTS
 Top
 Notes
 Abstract
 Introduction
 Materials and Methods
 Results
 Discussion
 References
 
Depsipeptide Treatment of Mice With EBV-Positive LCL and EBV-Positive Burkitt Lymphoma Tumors

To study the effects of depsipeptide in SCID mice, we first performed toxicity studies to determine the maximum tolerated dose and optimal treatment schedule. We evaluated daily injections of depsipeptide at 3, 5, and 10 mg/m2 and found substantial toxicity after a single dose at 10 mg/m2 or after three doses at 5 mg/m2. Rest periods of 2–3 days between doses at 5 mg/m2 reduced the toxicity substantially; thus, we defined 5 mg/m2 every other day to every third day as the maximum tolerated dose and treatment schedule. We next performed pharmacokinetic studies to determine plasma concentrations of depsipeptide achieved. After a single injection of depsipeptide at the maximum tolerated dose (5 mg/m2), the level of biologically active drug in plasma peaked at 15 minutes at 34 nM (95% CI = 0 to 70; n = 3 mice per sampling time) and then declined rapidly within 4 hours (Fig. 1, A).



View larger version (11K):
[in this window]
[in a new window]
 
Fig. 1. Depsipeptide and the survival of mice with Epstein-Barr virus (EBV)–positive type III latent (Lat-III) B-cell tumors and mice with EBV-positive type I latent (Lat-I) tumors. A) Pharmacokinetic analysis. Mice (five mice per group) received a single injection of depsipeptide (5 mg/m2 of body surface area), and plasma levels of depsipeptide were determined up to 8 hours after drug injection. Error bars = 95% confidence intervals [CIs]. B) Survival analysis. Mice (groups of 10) received four treatments of depsipeptide (5 mg/m2). For severe combined immune-deficient (SCID) mice engrafted with Lat-III human C7M3 EBV-positive LCLs, 90% (95% CI = 73.2% to 100%, 10 mice at risk) of the depsipeptide-treated mice remained alive at day 30, compared with 20% (95% CI = 5.8% to 69.1%, 10 mice at risk) of the mice treated with vehicle control. Ninety percent of depsipeptide-treated mice and 20% of the vehicle control–treated mice remained without tumor for 90 days. For mice engrafted with Raji cells, the survival probability was 30% (95% CI = 11.6% to 77.3%, 10 mice at risk) for depsipeptide-treated mice at day 22 and was 20% (95% CI = 5.79% to 69.1%, 10 mice at risk) for control-treated mice at day 20. All animals engrafted with Raji Burkitt lymphomas had become moribund with tumor burden and were removed from the study by day 23. Survival times were compared by use of the log-rank test. All statistical tests were two-sided.

 
When SCID mice engrafted with Lat-III human C7M3 EBV-positive LCLs were treated with depsipeptide (5 mg/m2) or vehicle control on days 2, 4, 7, and 10, 90% (95% CI = 73.2% to 100%; nine of 10 mice alive) of the mice treated with depsipeptide were alive at day 30, compared with 20% (95% CI = 5.8% to 69.1%; two of 10 mice alive) of the mice treated with vehicle control (Fig. 1, B; P<.001). No additional animals died in both depsipeptide- and control-treated groups engrafted with C7M3 LCLs when monitored for 90 days. The 20% survival seen in the vehicle control–treated group was likely due to a failure of tumor engraftment, a placebo treatment effect, or both. In contrast, we found essentially no difference in survival between depsipeptide-treated and control-treated mice engrafted with Lat-I human Raji EBV-positive Burkitt lymphoma cells. For mice engrafted with Raji cells, the survival probability at day 22 was 30% (95% CI = 11.6% to 77.3%, 10 mice at risk; three of 10 mice alive) for depsipeptide-treated mice, and the survival probability at day 20 was 20% (95% CI = 5.79% to 69.1%, 10 mice at risk; two of 10 mice alive) for control-treated mice. By day 23 of the trial, all mice engrafted with Raji Burkitt lymphoma tumors were found moribund from tumor burden, requiring removal from the study. Depsipeptide-treated mice engrafted with Lat-III C7M3 LCLs demonstrated a statistically significantly better survival at day 30 compared with depsipeptide-treated mice engrafted with Lat-I Raji Burkitt lymphoma cell lines (P<.001; Fig 1, B).

In humans, plasma concentrations of 450–1050 nM depsipeptide can be safely achieved over the course of a 4-hour intravenous infusion (4). Consequently, in our in vitro studies, we tested a biologically relevant dose range (50–500 nM depsipeptide) and exposure time (4 hours) that were based on the drug levels measured in SCID mice (Fig. 1, A) and the higher doses achieved in human subjects. Apoptosis was induced in 35%–64% of cells in a representative culture of depsipeptide-treated EBV-positive LCLs (PA-LCL; compared with 3%–6% of cells in a culture of vehicle-treated EBV-positive LCLs) after a single 4-hour treatment with 500 nM depsipeptide (Fig. 2, A). Similar results were obtained with C7M3 LCL and RM LCL. Depsipeptide induced apoptosis in EBV-positive LCLs in a dose-dependent fashion beginning at 10 nM in the C7M3, PA, and RM LCLs (data not shown). In contrast, treatment of EBV-positive or EBV-negative Burkitt lymphoma cell cultures with 500 nM depsipeptide resulted in a marginally higher percentage (14% and 12%, respectively) of apoptotic cells compared with that (3% and 6%, respectively) in vehicle-treated control cultures (Fig. 2, B and C).



View larger version (49K):
[in this window]
[in a new window]
 
Fig. 2. Depsipeptide, apoptosis, and proliferation of Epstein-Barr virus (EBV)–positive lymphoblastoid cell lines (LCLs) and EBV-negative and EBV-positive Burkitt lymphoma cell lines. AC) Cell viability and depsipeptide. A) Assessment of cell viability by flow cytometry with Annexin V–fluorescein isothiocyanate (FITC) and propidium iodide staining of EBV-positive LCLs 72 hours after a 4-hour 500 nM depsipeptide treatment. Percentages of viable, apoptotic, and necrotic cells are shown. Results from a single representative LCL (PA cells) are shown and are similar to those of two LCLs (C7M3 and RM) tested. B,C) Viability of EBV-positive and EBV-negative Burkitt lymphoma cell lines, respectively, were relatively unaffected by depsipeptide. DI) Cell proliferation. Bar + = cells incubated in standard growth medium; bar –= cells incubated with mitomycin C to inhibit incorporation of thymidine. Depsipeptide (500 nM) inhibited the proliferation (incorporation of [3H]thymidine, expressed as counts per minute [CPM]) of EBV-positive LCLs 72 hours after treatment (D); the proliferation of EBV-positive (Raji) and EBV-negative (BL41) Burkitt lymphoma cell lines, respectively, was unaffected (E,F). Absolute number of viable cells remained fixed in depsipeptide-treated EBV-positive LCL cultures (G), whereas the total number of viable cells in EBV-positive and EBV-negative Burkitt lymphoma cell cultures increased over a 96-hour period (H,I). Data in panels D–I represent the mean of three replicate experiments with 95% confidence intervals.

 
Twenty-four hours after EBV-positive LCLs (C7M3, PA, and RM) and Burkitt lymphoma cells (Raji, Daudi, and BL41) were treated for 4 hours with 500 nM depsipeptide, the level of total acetylation of H4 at lysine residues 5, 8, 12, and 16 was higher than that in corresponding untreated control cells; the level of acetylation subsequently returned to near-baseline levels by 48 hours. Depsipeptide treatment of peripheral blood mononuclear cells from two normal human donors resulted in a similar increase in H4 acetylation (data not shown). In addition, EBV-positive LCLs (C7M3, PA, and RM) treated with depsipeptide for 4 hours stopped replicating DNA and proliferating, as measured by [3H]thymidine incorporation (Fig. 2, D), whereas Burkitt lymphoma cells treated with depsipeptide for 4 hours continued to replicate DNA and proliferate (Fig. 2, E and F). Absolute cell numbers determined 96 hours after treatment with depsipeptide further supported these data (Fig. 2, G–I).

Induction of Apoptosis in EBV-Positive LCLs with Depsipeptide and Other Histone Deacetylase Inhibitors

To compare the proapoptotic activity of the depsipeptide with other compounds with histone deacetylase inhibitor activity, we performed dose-escalation experiments with trichostatin A and butyrate. Dose-escalation experiments with a 4-hour exposure demonstrated that trichostatin A was cytotoxic at concentrations 600-fold greater than depsipeptide and that butyrate was cytotoxic at concentrations 600 000-fold greater than depsipeptide (Fig. 3). In contrast to depsipeptide, however, both compounds were equally cytotoxic to cell lines with Lat-I (Raji) and Lat-III (C7M3, PA, and RM) viral gene expression profiles (data not shown).



View larger version (56K):
[in this window]
[in a new window]
 
Fig. 3. Induction of apoptosis in Epstein-Barr virus (EBV)–positive lymphoblastoid cell lines (LCLs) with depsipeptide and other histone deacetylase inhibitors. Cells were stained with Annexin V and propidium iodide to assess percentage of viable, apoptotic, and necrotic cells via flow cytometry. Substantially higher concentrations of the histone deacetylase inhibitors butyrate (3 x 105 µM; C) and trichostatin A (TSA, 3 x 102 µM; B) than of depsipeptide (5 x 10–2 µM; A) were required to induce apoptosis 72 hours after a 4-hour treatment. Data shown are derived from experiments with C7M3 LCL and are representative of three different EBV-positive LCLs (C7M3, PA LCL, and RM LCL).

 
Depsipeptide-Induced Apoptosis and the Activation of Caspase 3

A major pathway leading to apoptosis involves activation of multiple caspase proteins that converge on the cleavage and subsequent activation of caspase 3, which in turn triggers other apoptotic cascades. Treatment of the three EBV-positive LCLs (C7M3, PA, and RM) with depsipeptide induced statistically significantly more active caspase 3 after 48 hours than that in corresponding control cultures treated with vehicle (Fig. 4, A; P<.001). The percentage of cells staining positive for active caspase 3 (Fig. 4, A, lower panel) was associated with levels of apoptosis as determined by Annexin V and propidium iodide staining (Fig. 2, A) and by nuclear DNA staining (Fig. 4, B, right panel). Cells in which active caspase 3 was not detected also had a normal DNA content, with the proportions of cells in G1, S, and G2/M phases expected for proliferating cells (Fig. 4, B, left panel). Inhibition of caspase 3 with the broad-spectrum caspase inhibitor zVAD-fmk before treatment with depsipeptide partially reduced depsipeptide-induced apoptosis from 90% of cells in apoptosis (Fig. 5, A, no zVAD-fmk treatment) to 63% (Fig. 5, B, addition of zVAD-fmk). Western blot analysis for the active form of caspase 3 demonstrated that the broad-spectrum caspase inhibitor zVAD-fmk completely inhibited caspase 3 cleavage (i.e., activation) in EBV-positive LCLs treated with anti-Fas or depsipeptide (Fig. 5, C). These results demonstrate that depsipeptide-induced apoptosis of EBV-positive LCLs occurs via mechanisms that are caspase dependent and mechanisms that are caspase independent.



View larger version (24K):
[in this window]
[in a new window]
 
Fig. 4. Depsipeptide, activation of caspase 3, and apoptotic DNA fragmentation in Epstein-Barr virus (EBV)–positive lymphoblastoid cell lines (LCLs). A) Intracellular flow cytometry. (Upper) EBV-positive LCLs treated with vehicle control displayed little active caspase 3 staining (Casp-3POS). (Lower) Forty-eight hours after a 4-hour treatment with 500 nM depsipeptide, 60.5% of cells had increased levels of active caspase 3 staining, but 39.5% of cells still lacked active caspase 3. B) EBV-positive LCLs treated with 500 nM depsipeptide were gated (i.e., separated) as either lacking active caspase 3 (Caspase 3NEG) or expressing active caspase 3 (Caspase-3POS) and analyzed for cell cycle position. Cells that lacked active caspase 3 (Caspase-3NEG; left panel) had a normal cell cycle distribution and no DNA degradation, whereas cells with active caspase 3 (Caspase-3POS; right panel) had substantial DNA degradation, characteristic of apoptosis or necrosis. Data shown are derived from experiments with PA LCL and are representative of three different EBV-positive LCLs (C7M3, PA LCL, and RM LCL).

 


View larger version (57K):
[in this window]
[in a new window]
 
Fig. 5. Depsipeptide-induced cell death in Epstein-Barr virus (EBV)–positive type III latent lymphoblastoid cell lines (LCLs) and broad-spectrum inhibition of caspases. A) Treatment with depsipeptide alone. Depsipeptide treatment (100 nM) of EBV-positive C7M3 LCLs resulted in apoptosis after 48 hours. B) Pretreatment with 50 µM z-VAD-fmk followed by treatment with 100 nM depsipeptide resulted in reduced apoptosis after 48 hours. A, B) Data are representative of three EBV-positive LCLs (C7M3, PA, and RM). C) Immunoblot analysis of cleaved caspase 3. Active caspase 3 has a molecular mass of 12 kd. Pretreatment of C7M3 LCLs with z-VAD-fmk inhibits caspase enzymatic activity. The 12-kd active caspase 3 is induced in cells treated with the agonistic anti-Fas monoclonal antibody or depsipeptide (lanes 2 and 7). Pretreatment with 10 µM z-VAD-fmk prevents the cleavage of active caspase 3 and is, therefore, an indication that caspase activity is inhibited (lanes 5, 8, and 9). Blot is representative of results with two separate EBV-positive LCLs, C7M3 and RM.

 
LMP1 and BCL2 in Depsipeptide-Induced Apoptosis

We consistently observed a small subset of Lat-III EBV–positive LCLs that remained viable despite treatment with high concentrations of depsipeptide. In repeated experiments, viability was not dependent on phase of the cell cycle (data not shown). Tumor cells resistant to depsipeptide-mediated apoptosis could be clearly identified by flow cytometry as a population that was negative for Annexin V binding, propidium iodide binding, and active caspase 3 (Figs. 2 and 4, respectively). Because the LMP1 protein is critical to survival of EBV-positive LCLs (19,20), we used intracellular flow cytometry to compare LMP1 levels in depsipeptide-sensitive and -resistant populations. Forty-eight hours after treatment with depsipeptide, cells were separated by flow cytometry into an active caspase 3–positive population or an active caspase 3–negative population, and then LMP-1 expression was evaluated in each population (Fig. 6, A). Subpopulations with low and high LMP1 expression were present before depsipeptide treatment. This heterogeneity in LMP1 expression resulted in a mean fluorescent intensity (MFI) of 39.96 U (Fig. 6, B). Forty-eight hours after a 4-hour exposure to depsipeptide, cells with active caspase 3 were predominantly those expressing low levels of LMP1 (MFI = 13.13 U, Fig. 6, C), whereas cells lacking active caspase 3 expressed higher levels of LMP1 (MFI = 61.77 U, Fig. 6, D) (P = .017). The same pattern was true for the antiapoptotic protein BCL2 (Fig. 6, D; P = .032), a downstream target of LMP1 (20). Thus, expression of LMP1 and BCL2 was associated with caspase 3 activation after treatment with depsipeptide. Given the ability of LMP1 to trigger the activation of several cellular survival pathways, the mechanisms by which EBV-positive LCLs survive depsipeptide treatment likely involve one or more downstream targets of LMP1.



View larger version (34K):
[in this window]
[in a new window]
 
Fig. 6. Caspase 3 activity in Epstein-Barr virus (EBV)–positive lymphoblastoid cell lines (LCLs) expressing high levels of LMP1 and BCL2 protein. A) Forty-eight hours after a 4-hour treatment with either depsipeptide or vehicle control, EBV-positive LCLs were evaluated for intracellular active caspase 3, LMP1, and BCL2. Casp-3NEG = cells staining negative for active caspase 3; Casp-3POS = cells staining positive for active caspase 3. B) Intracellular expression of LMP1 and BCL2 protein. Baseline expression of LMP1 (left) and BCL2 (right) protein in untreated EBV-positive LCLs shows pretreatment heterogeneity of intracellular protein expression. Depsipeptide-treated (500 nM) EBV-positive LCLs were divided into a population of cells expressing active caspase 3 (Casp-3POS; C) and a population of cells not expressing active caspase 3 (Casp-3NEG; D) and then evaluated for expression of LMP1 (left) and BCL2 (right). Cells lacking active caspase 3 had a higher mean fluorescence intensity (MFI) for LMP1 (P = .017) and BCL2 (P = .032) than cells with active caspase 3. Data shown are derived from experiments with PA LCL and are representative of three EBV-positive LCLs (C7M3, PA, and RM). All statistical tests were two-sided.

 
Specific Inhibition of NF-{kappa}B and Depsipeptide-Induced Apoptosis

LMP1 protein can also enhance the survival and growth of EBV-transformed lymphoblasts by constitutive activation of the transcription factor NF-{kappa}B (21). To investigate whether this potential survival mechanism was operating in depsipeptide-resistant cells, we isolated viable cells (LMP1-positive, BCL2-positive, and caspase 3-negative cells) that survived a high-dose (500 nM) depsipeptide treatment and evaluated their NF-{kappa}B activity. These depsipeptide-resistant cells exhibited higher levels of NF-{kappa}B p65–containing dimer than cells treated with vehicle control (absorbance at 450 nm = 1.57 versus 1.13; P<.001) (Fig. 7, A).



View larger version (17K):
[in this window]
[in a new window]
 
Fig. 7. Nuclear factor {kappa}B (NF-{kappa}B) activity and depsipeptide-induced apoptosis. A) NF-{kappa}B activity. NF-{kappa}B activity was assayed using the NF-{kappa}B Trans-Am kit. (–)Ctrl = tumor necrosis factor {alpha} (TNF-{alpha})–treated HeLa cell extracts incubated with excess wild-type NF-{kappa}B–specific oligonucleotide; (+)Ctrl = TNF-{alpha}–treated HeLa cell extracts incubated with excess mutant NF-{kappa}B–specific oligonucleotide; Depsi = depsipeptide. B) Induction of apoptosis with NF-{kappa}B inhibition. Apoptosis was assessed with Annexin V and propidium iodide staining. Data are expressed as means of three separate experiments with three separate LCLs (C7M3, PA, and RM) and upper 95% confidence intervals. All statistical tests were two-sided.

 
Because EBV-positive LCLs that were resistant to depsipeptide expressed relatively high levels of LMP1 and its downstream transcription factor NF-{kappa}B, we next determined whether specifically inhibiting NF-{kappa}B could enhance depsipeptide-induced apoptosis. Inhibition of NF-{kappa}B with PS-1145 alone had little effect on EBV-positive LCL viability (Fig. 7, B). However, in three EBV-positive LCLs tested, specific inhibition of NF-{kappa}B resulted in a statistically significant increase in depsipeptide-induced apoptosis (P<.001; Fig. 7, B). Treatment of Lat-I cell lines with PS-1145 did not affect cell viability 48 hours after treatment (data not shown). Inhibition of NF-{kappa}B with the selective inhibitor PS1145 promotes depsipeptide-induced apoptosis and indicates that key downstream targets of LMP1 important to survival mechanisms are operable in EBV-positive LCLs.


    DISCUSSION
 Top
 Notes
 Abstract
 Introduction
 Materials and Methods
 Results
 Discussion
 References
 
In this study, we present preclinical data on the selective antitumor activity of depsipeptide against EBV-positive lymphoma cell lines that have a Lat-III viral gene profile. Single-agent depsipeptide therapy improved the long-term survival of SCID mice engrafted with an established Lat-III EBV–positive LCL tumor but not of SCID mice engrafted with Lat-I EBV–positive Burkitt lymphoma tumors. This effect was selective for depsipeptide and occurred at concentrations and exposure times that are far below the maximum tolerated dose identified in humans (4). Our in vitro characterization showed that depsipeptide treatment of EBV-positive LCLs induced apoptosis at concentrations (45–1000 nM) and exposure times (4 hours) achievable in humans (4). The induction of apoptosis was not associated with the cell’s phase of the cell cycle and involved caspase-dependent and -independent mechanisms. Endogenous survival pathways driven by the EBV-encoded LMP1 protein likely account for the heterogeneous in vitro response to depsipeptide and could be partially circumvented by inhibiting constitutive activation of NF-{kappa}B with the I{kappa}K inhibitor PS-1145.

As with chromatin in the cell’s nucleus, the EBV episome is organized into a tightly compacted nucleosomal structure that requires multisubunit enzymatic complexes to alter its nucleosomal conformation and allow coordinated gene expression. The regulation of latent and lytic EBV gene expression is complex and requires both covalent (2225) and ATP-dependent (26,27) mechanisms to modulate promoter accessibility and thus the expression of a specific latent (Lat-I, -II, or -III) or lytic gene profile (2830). It is possible that the antiproliferative effect of depsipeptide in EBV-positive LCLs (Fig. 2) involves activation of lytic transcription, because we have observed an induction of intermediate early lytic gene products after exposure of EBV-positive LCLs to depsipeptide (data not shown) (31). Unlike other histone deacetylase inhibitors that we examined (trichostatin A and butyrate), depsipeptide did not have antiproliferative, proapoptotic activity on Burkitt lymphoma cell lines. Although the mechanism for this apparent selective death of Lat-III EBV-positive LCLs is not clear, depsipeptide-mediated histone deacetylase inhibition may derepress Myc target genes, as described previously by Pal et al. (32). This activity could then enhance the proliferation and survival of EBV-positive Burkitt lymphoma cell lines that possess the IgH–c-myc translocation, which drives the constitutive expression of Myc.

Alternatively, depsipeptide may be exerting these selective properties through a mechanism that is independent of histone acetylation. Gu et al. (33) reported that p53 DNA binding affinity is regulated by acetylation of lysine residues in the carboxyl terminus of p53. Treatment with a histone deacetylase inhibitor can result in the acetylation of specific lysine residues in p53, leading to the increased stability and transcription of p53-driven proapoptotic target genes (34,35). Because 60%–80% of Burkitt lymphoma cell lines have mutations in p53, this may render them more tumorigenic in vivo and possibly resistant to the effects of acetylation mediated by histone deacetylase inhibitor treatment (36).

We observed a 12-fold increase in the level of active caspase 3 after cells were briefly exposed to depsipeptide. Pretreatment of EBV-positive LCLs with z-VAD-fmk resulted in complete inhibition of caspase 3 cleavage, as detected by western blot analysis, but in only partial inhibition of depsipeptide-induced apoptosis, as determined by flow cytometry with Annexin and propidium iodide staining. Susin et al. (37) reported that apoptosis-inducing factor is a mediator of caspase-independent apoptosis. Although pretreatment of EBV-positive LCLs with the broad-spectrum caspase inhibitor z-VAD-fmk partially inhibited depsipeptide-induced cell death, it effectively prevented high-molecular-weight DNA fragmentation, a characteristic feature of apoptosis induced by apoptosis-inducing factor (data not shown). Thus, it appears that the mechanism of depsipeptide-mediated apoptosis in EBV-positive LCLs is independent of caspase 3 and apoptosis-inducing factor (37).

Despite depsipeptide’s induction of apoptosis, we observed a subset of cells that were resistant to depsipeptide-induced apoptosis in vitro. This heterogeneity may reflect heterogeneous expression of the Lat-III–associated viral gene product LMP1 in EBV-positive LCLs (38). LMP1, a member of the tumor necrosis factor receptor family, uses tumor necrosis factor receptor–associated factors to bypass CD40 signaling leading to the constitutive activation of the survival factors BCL2 and NF-{kappa}B in EBV-transformed lymphoblasts (19,39,40). We observed a similar degree of heterogeneity of LMP1 expression with both untreated and depsipeptide-treated EBV-positive LCLs; thus, it is possible that EBV-positive LCLs that express high levels of LMP1 and the downstream survival mediators NF-{kappa}B and BCL2 would selectively survive exposure to depsipeptide.

It is also possible that depsipeptide-mediated histone deacetylase inhibition leads to changes in LMP1 expression via epigenetic mechanisms. Indeed, several reports have described that histone deacetylase and histone acetyltransferase activity regulate the LMP1 promoter (2426,41,42). In addition, Sjoblom-Hallen et al. (43) demonstrated that expression of LMP1 is silenced by a multisubunit complex containing Max, Mad1, mSin3A, and histone deacetylase that localizes to an E-box site present within the LMP1 promoter. EBNA3C, an EBV transcription factor belonging to the Lat-III viral gene profile, recruits histone deacetylases and represses EBNA2-activated transcription, including that at the LMP1 promoter (24,42). By the use of chromatin immunoprecipitation experiments with monoclonal antibodies specific for EBNA2 and acetylated histones, Alazard et al. (41) showed that the EBV-encoded coactivator EBNA2 binds to the LMP1 promoter and that LMP1 promoter–associated histone H3 acetylation is linked with recruitment of EBNA2. Furthermore, Park et al. (38) showed that treatment with nontoxic doses of the histone deacetylase inhibitors butyrate (2.5 x 103 µM) and trichostatin A (0.332 µM) can induce the expression of LMP1 mRNA and protein in EBV-positive Burkitt lymphomas. To address whether depsipeptide could be having the same effect, we isolated the population of EBV-positive LCLs expressing high levels of LMP1 (LMP1high) and the population of EBV-positive LCLs expressing low levels of LMP1 (LMP1l°w) by flow cytometric sorting of distinct populations of LCLs with high or low CD54 surface expression, as described by Park et al. (38). We found that depsipeptide treatment of EBV-positive LCLs with low CD54 expression (also LMP1l°w) enhanced the expression of LMP1 (data not shown). Thus, depsipeptide-mediated inhibition of histone deacetylase appears to simultaneously induce apoptosis and LMP1 expression, leading to the downstream activation of key survival pathways, including the LMP1 targets BCL2 and NF-{kappa}B (44).

LMP1 signaling increases the expression of BCL2 in EBV-infected B cells (20). We found an association between survival and the expression of LMP1 and BCL2 in depsipeptide-treated cells (Fig. 6). BCL2 can inhibit both caspase-dependent and -independent apoptosis by limiting the permeability of the mitochondrial membrane, thereby preventing the contents of the mitochondrial matrix from reaching the cytosol. In particular, BCL2 can prevent the release and subsequent activation of caspase-dependent proapoptotic factors, including cytochrome c and Bid (45). An earlier study (40) reported that treatment of EBV-positive LCLs with LMP1 antisense RNA decreased BCL2 levels, reduced proliferative capacity, and induced apoptosis under serum-free conditions. Thus, LMP1-mediated induction of BCL2 would lead to resistance of both caspase-dependent and -independent apoptosis after exposure to depsipeptide.

A second element of LMP1 signal transduction that may contribute to depsipeptide resistance is the constitutive activation of the transcription factor NF-{kappa}B. LMP1 constitutively activates NF-{kappa}B in EBV-positive LCLs through tumor necrosis factor receptor–associated factors, which activate NF-{kappa}B–inducing kinases and I{kappa}B kinases to phosphorylate I{kappa}B{alpha}, thus promoting I{kappa}B{alpha} degradation by the proteasome (46). I{kappa}B{alpha} prevents NF-{kappa}B dimerization and binding to target sequences within DNA by binding and sequestering NF-{kappa}B subunits in the cytoplasm. Viable EBV-positive LCLs that persisted after depsipeptide treatment had higher levels of NF-{kappa}B activity than untreated control EBV-positive LCLs (Fig. 7). Thus, surviving cells may have responded to depsipeptide treatment by increasing their NF-{kappa}B activity. To further explore the contribution of NF-{kappa}B to this apparent intrinsic resistance to depsipeptide, we inhibited LMP1-mediated activation of NF-{kappa}B with PS-1145, a specific inhibitor of I{kappa}B kinase (17). Pretreatment of cultures with PS-1145 alone had no effect on cell viability, but combined treatment of cultures with PS-1145 and depsipeptide resulted in increased apoptosis compared with cultures treated with depsipeptide alone. Because preexisting NF-{kappa}B activity is not affected by PS-1145, there was only partial inhibition of LMP1-driven NF-{kappa}B activation. Expression of a dominant negative NF-{kappa}B in EBV-positive LCLs has been reported to cause spontaneous apoptosis (47). It is possible that partial inhibition of NF-{kappa}B activity lowered the apoptotic threshold of EBV-positive LCLs, lending support to the hypothesis that NF-{kappa}B is a resistance factor against depsipeptide-induced apoptosis. Indeed, recent work has identified binding sites for several components of NF-{kappa}B complexes within the BCL2 (48) and A1/Bfl1 (49) promoters, suggesting that important survival gene products are downstream targets of NF-{kappa}B activity.

With the more frequent use of stem cell and solid organ transplantation and the accelerating worldwide AIDS epidemic, the number of individuals with chronic immune suppression will continue to increase. Thus, it is vital to continue development of novel strategies that effectively target virally associated opportunistic neoplasms in immune-compromised individuals. In an era where drug development has shifted toward molecular targeting, reagents with histone deacetylase inhibitor activity are emerging as attractive compounds to test in clinical trials. Compounds with histone deacetylase inhibitor activity induce growth arrest, differentiation, and apoptosis in various tumor cell lines in vitro and in several preclinical animal models of cancer (3). Although the development of many of these compounds has been limited by their suboptimal pharmacokinetics and high toxicity profiles (50), depsipeptide is a potent histone deacetylase inhibitor that can reach plasma concentrations of 400–1000 nM in humans and has a proven safety record in phase I trials (4,5). Although the potential of depsipeptide as a treatment for EBV-associated B-cell malignancies has yet to be explored clinically, the in vivo and in vitro data from this study suggest that use of depsipeptide as single agent, followed by a combination treatment with depsipeptide and PS-1145, would be a reasonable approach to the treatment of EBV-associated B-cell lymphoproliferative disorders.


    NOTES
 Top
 Notes
 Abstract
 Introduction
 Materials and Methods
 Results
 Discussion
 References
 
S. Roychowdhury and R. A. Baiocchi contributed equally to this work.

Supported by National Institutes of Health grant T32 CA09338 to R. A. Baiocchi and by National Institutes of Health grant P01 CA95426 to C. F. Eisenbeis and M. A. Caligiuri.

We thank Millennium Pharmaceuticals, Inc., for PS-1145 compound. We also thank John Wright from the National Cancer Institute and Fujisawa Pharmaceuticals, Inc., for providing depsipeptide. We thank Donna Bucci, Tamra Brooks, and Karen Feasel for administrative assistance and preparation/submission of the manuscript.


    REFERENCES
 Top
 Notes
 Abstract
 Introduction
 Materials and Methods
 Results
 Discussion
 References
 

1 Rickinson AB, Kieff E. Epstein-Barr virus. In: Fields BN, editor. Fields’ virology. Vol 1. 4th ed. New York (NY): Lippincott Williams & Wilkins; 2001. p. 2575–627.

2 Kieff E, Rickinson AB. Epstein-Barr virus and replication. In Fields BN, editor. Fields’ virology. Vol 1. 4th ed. New York (NY): Lippincott Williams & Wilkins; 2001. p. 2511–73.

3 Johnstone RW. Histone-deacetylase inhibitors: novel drugs for the treatment of cancer. Nat Rev Drug Discov 2002;1:287–99.[CrossRef][ISI][Medline]

4 Sandor V, Bakke S, Robey RW, Kang MH, Blagosklonny MV, Bender J, et al. Phase I trial of the histone deacetylase inhibitor, depsipeptide (FR901228, NSC 630176), in patients with refractory neoplasms. Clin Cancer Res 2002;8:718–28.[Abstract/Free Full Text]

5 Piekarz RL, Robey R, Sandor V, Bakke S, Wilson WH, Dahmoush L, et al. Inhibitor of histone deacetylation, depsipeptide (FR901228), in the treatment of peripheral and cutaneous T-cell lymphoma: a case report. Blood 2001;98:2865–8.[Abstract/Free Full Text]

6 Baiocchi RA, Caligiuri MA. Low-dose interleukin 2 prevents the development of Epstein-Barr virus (EBV)-associated lymphoproliferative disease in scid/scid mice reconstituted i.p. with EBV-seropositive human peripheral blood lymphocytes. Proc Natl Acad Sci U S A 1994;91:5577–81.[Abstract]

7 Tanaka T, Kitamura F, Nagasaka Y, Kuida K, Suwa H, Miyasaka M. Selective long-term elimination of natural killer cells in vivo by an anti-interleukin 2 receptor beta chain monoclonal antibody in mice. J Exp Med 1993;178:1103–7.[Abstract]

8 Li Z, Chan KK. A subnanogram API LC/MS/MS quantitation method for depsipeptide FR901228 and its preclinical pharmacokinetics. J Pharm Biomed Anal 2000;22:33–44.[CrossRef][ISI][Medline]

9 Vermes I, Haanen C, Steffens-Nakken H, Reutelingsperger C. A novel assay for apoptosis. Flow cytometric detection of phosphatidylserine expression on early apoptotic cells using fluorescein labelled Annexin V. J Immunol Methods 1995;184:39–51.[CrossRef][ISI][Medline]

10 Cooper MA, Fehniger TA, Turner SC, Chen KS, Ghaheri BA, Ghayur T, et al. Human natural killer cells: a unique innate immunoregulatory role for the CD56(bright) subset. Blood 2001;97:3146–51.[Abstract/Free Full Text]

11 Baiocchi RA, Ross ME, Tan JC, Chou CC, Sullivan L, Haldar S, et al. Lymphomagenesis in the SCID-hu mouse involves abundant production of human interleukin-10. Blood 1995;85:1063–74.[Abstract/Free Full Text]

12 Robertson MJ, Manley TJ, Pichert G., Cameron C, Cochran KJ, Levine H, et al. Functional consequences of APO-1/Fas (CD95) antigen expression by normal and neoplastic hematopoietic cells. Leuk Lymphoma 1995;17:51–61.[ISI][Medline]

13 Nicoletti I, Migliorati G, Pagliacci MC, Grignani F, Riccardi C. A rapid and simple method for measuring thymocyte apoptosis by propidium iodide staining and flow cytometry. J Immunol Methods 1991;139:271–9.[CrossRef][ISI][Medline]

14 Susin SA, Daugas E, Ravagnan L, Samejima K, Zamzami N, Loeffler M, et al. Two distinct pathways leading to nuclear apoptosis. J Exp Med 2000;192:571–80.[Abstract/Free Full Text]

15 Zhu J, Song X, Lin HP, Young DC, Yan S, Marquez VE, et al. Using cyclooxygenase-2 inhibitors as molecular platforms to develop a new class of apoptosis-inducing agents. J Natl Cancer Inst 2002;94:1745–57.[Abstract/Free Full Text]

16 Aron JL, Parthun MR, Marcucci G, Kitada S, Mone AP, Davis ME, et al. Depsipeptide (FR901228) induces histone acetylation and inhibition of histone deacetylase in chronic lymphocytic leukemia cells concurrent with activation of caspase 8-mediated apoptosis and down-regulation of c-FLIP protein. Blood 2003;102:652–8.[Abstract/Free Full Text]

17 Hideshima T, Chauhan D, Richardson P, Mitsiades C, Mitsiades N, Hayashi T, et al. NF-kappa B as a therapeutic target in multiple myeloma. J Biol Chem 2002;277:16639–47.[Abstract/Free Full Text]

18 Klein JP, Moeschberger, ML. Survival analysis: techniques for censored and truncated data. New York (NY): Springer; 1997.

19 Gregory CD, Dive C, Henderson S, Smith CA, Williams GT, Gordon J, et al. Activation of Epstein-Barr virus latent genes protects human B cells from death by apoptosis. Nature 1991;349:612–4.[CrossRef][ISI][Medline]

20 Henderson S, Rowe M, Gregory C, Croom-Carter D, Wang F, Longnecker R, et al. Induction of bcl-2 expression by Epstein-Barr virus latent membrane protein 1 protects infected B cells from programmed cell death. Cell 1991;65:1107–15.[ISI][Medline]

21 Devergne O, Hatzivassiliou E, Izumi KM, Kaye KM, Kleijnen MF, Kieff E, et al. Association of TRAF1, TRAF2, and TRAF3 with an Epstein-Barr virus LMP1 domain important for B-lymphocyte transformation: role in NF-kappaB activation. Mol Cell Biol 1996;16:7098–108.[Abstract]

22 Chang L, Liu ST. Activation of the BRLF1 promoter and lytic cycle of Epstein-Barr virus by histone acetylation. Nucleic Acids Res 2000;28:3918–25.[Abstract/Free Full Text]

23 Jenkins PJ, Binne UK, Farrell PJ. Histone acetylation and reactivation of Epstein-Barr virus from latency. J Virol 2000;74:710–20.[Abstract/Free Full Text]

24 Radkov S, Touitou R, Brehm A, Rowe M, West M, Kourzarides R, et al. Epstein-Barr virus nuclear antigen 3C interacts with histone deacetylase to repress transcription. J Virol 1999;73:5688–97.[Abstract/Free Full Text]

25 Wang L, Grossman SR, Kieff E. Epstein-Barr virus nuclear protein 2 interacts with p300, CBP, and PCAF histone acetyltransferases in activation of the LMP1 promoter. Proc Natl Acad Sci U S A 2000;97:430–5.[Abstract/Free Full Text]

26 Sjoblom-Hallen A, Yang W, Jansson A, Rymo L. Silencing of the Epstein-Barr virus latent membrane protein 1 gene by the Max-Mad1-mSin3A modulator of chromatin structure. J Virol 1999;73:2983–93.[Abstract/Free Full Text]

27 Wu DY, Kalpana GV, Goff SP, Schubach WH. Epstein-Barr virus nuclear protein 2 (EBNA2) binds to a component of the human SNF-SWI complex, hSNF5/Ini1. J Virol 1996;70:6020–8.[Abstract]

28 Knight JS, Lan K, Subramanian C, Robertson ES. Epstein-Barr virus nuclear antigen 3C recruits histone deacetylase activity and associates with the corepressors mSin3A and NCoR in human B cell lines. J Virol 2003;77:4261–72.[Abstract/Free Full Text]

29 Corcoran EE, Means AR. Defining Ca(2+)/calmodulin-dependent protein kinase cascades in transcriptional regulation. J Biol Chem 2002;276:2975–8.[CrossRef][ISI]

30 Blaeser F, Ho N, Prywes R, Chatila TA. Ca(2+)-dependent gene expression mediated by MEF2 transcription factors. J Biol Chem 2000;275:197–209.[Abstract/Free Full Text]

31 Baiocchi RA, Sekula T, Vourganti S, Nair V, Roychowdhury S, Parthun M, et al. Treatment of EBV-transformed B cell lines with depsipeptide results in the induction of a viral thymidine kinase and enhanced susceptibility to antiviral-induced apoptosis. Proceedings of the EORTC, NCI, and American Association for Cancer Research; 2001 Nov 2; Miami, FL.

32 Pal S, Yun R, Datta A, Lacomis L, Erdjument-Bromage H, Kumar J, et al. mSin3A/histone deacetylase 2- and PRMT5-containing Brg1 complex is involved in transcriptional repression of the Myc target gene cad. Mol Cell Biol 2003;23:7475–87.[Abstract/Free Full Text]

33 Gu W, Roeder RG. Activation of p53 sequence-specific DNA binding by acetylation of the p53 C-terminal domain. Cell 1997;90:595–606.[ISI][Medline]

34 Terui T, Murakami K, Takimoto R, Takahashi M, Takada K, Murakami T, et al. Induction of PIG3 and NOXA through acetylation of p53 at 320 and 373 lysine residues as a mechanism for apoptotic cell death by histone deacetylase inhibitors. Cancer Res 2003;63:8948–54.[Abstract/Free Full Text]

35 Li M, Luo J, Brooks CL, Gu W. Acetylation of p53 inhibits its ubiquitination by Mdm2. J Biol Chem 2002;277:50607–11.[Abstract/Free Full Text]

36 Cherney BW, Bhatia KG, Sgadari C, Gutierrez MI, Mostowski H, Pike SE, et al. Role of the p53 tumor suppressor gene in the tumorigenicity of Burkitt’s lymphoma cells. Cancer Res 1997;57:2508–15.[Abstract]

37 Susin SA, Lorenzo HK, Zamzami N, Marzo I, Snow BE, Brothers GM, et al. Molecular characterization of mitochondrial apoptosis-inducing factor. Nature 1999;397:441–6.[CrossRef][ISI][Medline]

38 Park JH, Faller DV. Epstein-Barr virus latent membrane protein-1 induction by histone deacetylase inhibitors mediates induction of intercellular adhesion molecule-1 expression and homotypic aggregation. Virology 2002;303:345–63.[CrossRef][ISI][Medline]

39 Mosialos G. Cytokine signaling and Epstein-Barr virus-mediated cell transformation. Cytokine Growth Factor Rev 2001;12:259–70.[CrossRef][ISI][Medline]

40 Kenney JL, Guinness ME, Reiss M, Lacy J. Antisense to the Epstein-Barr virus (EBV)-encoded latent membrane protein 1 (LMP-1) sensitizes EBV-immortalized B cells to transforming growth factor-beta and chemotherapeutic agents. Int J Cancer 2001;91:89–98.[CrossRef][ISI][Medline]

41 Alazard N, Gruffat H, Hiriart E, Sergeant A, Manet E. Differential hyperacetylation of histones H3 and H4 upon promoter-specific recruitment of EBNA2 in Epstein-Barr virus chromatin. J Virol 2003;77:8166–72.[Abstract/Free Full Text]

42 Knight JS, Lan K, Subramanian C, Robertson ES. Epstein-Barr virus nuclear antigen 3C recruits histone deacetylase activity and associates with the corepressors mSin3A and NCoR in human B-cell lines. J Virol 2003;77:4261–72.[Abstract/Free Full Text]

43 Sjoblom-Hallen A, Yang W, Jansson A, Rymo L. Silencing of the Epstein-Barr virus latent membrane protein-1 gene by the Max-Mad-mSin3A modulator of chromatin structure. J Virol 1999;73:2983–93.[Abstract/Free Full Text]

44 Fries KL, Miller WE, Raab-Traub N. Epstein-Barr virus latent membrane protein 1 blocks p53 mediated apoptosis through the induction of the A20 gene. J Virol 1996;70:8653–9.[Abstract]

45 Cory S, Huang DC, Adams JM. The Bcl-2 family: roles in cell survival and oncogenesis. Oncogene 2003;22:8590–607.[CrossRef][ISI][Medline]

46 Cahir McFarland ED, Izumi KM, Mosialos G. Epstein-Barr virus transformation: involvement of latent membrane protein 1-mediated activation of NF-kappaB. Oncogene 1999;18:6959–64.[CrossRef][ISI][Medline]

47 Cahir-McFarland ED, Davidson DM, Schauer SL, Duong J, Kieff E. NF-kappa B inhibition causes spontaneous apoptosis in Epstein-Barr virus-transformed lymphoblastoid cells. Proc Natl Acad Sci U S A 2000;97:6055–60.[Abstract/Free Full Text]

48 Catz SD, Johnson JL. Transcriptional regulation of bcl-2by nuclear factor kB and its significance in prostate cancer. Oncogene 2001;20:7342–51.[CrossRef][ISI][Medline]

49 Wang CY, Guttridge DC, Mayo M, Baldwin AS. NF-kappaB induces expression of the Bcl2 homologue A1/BfL-1 to preferentially suppress chemotherapy-induced apoptosis. Mol Cell Biol 1999;19:5923–9.[Abstract/Free Full Text]

50 Miller AA, Kurschel E, Osieka R, Schmidt CG. Clinical pharmacology of sodium butyrate in patients with acute leukemia. Eur J Cancer Clin Oncol 1987;23:1283–7.[ISI][Medline]

Manuscript received January 23, 2004; revised July 29, 2004; accepted August 15, 2004.


This article has been cited by other articles in HighWire Press-hosted journals:


             
Copyright © 2004 Oxford University Press (unless otherwise stated)
Oxford University Press Privacy Policy and Legal Statement