Purpose: Histone deacetylase (HDAC) inhibition causes transcriptional activation or repression of several genes that in turn can influence the biodistribution of other chemotherapeutic agents. Here, we hypothesize that the combination of vorinostat, a HDAC inhibitor, with 131I-meta-iodobenzylguanidine (MIBG) would lead to preferential accumulation of the latter in neuroblastoma (NB) tumors via increased expression of the human norepinephrine transporter (NET).

Experimental Design:In vitro and in vivo experiments examined the effect of vorinostat on the expression of NET, an uptake transporter for 131I-MIBG. Human NB cell lines (Kelly and SH-SY-5Y) and NB1691-luc mouse xenografts were employed. The upregulated NET protein was characterized for its effect on 123I-MIBG biodistribution.

Results: Preincubation of NB cell lines, Kelly, and SH-SY-5Y, with vorinostat caused dose-dependent increases in NET mRNA and protein levels. Accompanying this was a corresponding dose-dependent increase in MIBG uptake in NB cell lines. Four- and 2.5-fold increases were observed in Kelly and SH-SY-5Y cells, respectively, pretreated with vorinostat in comparison to untreated cells. Similarly, NB xenografts, created by intravenous tail vein injection of NB1691-luc, and harvested from nude mice livers treated with vorinostat (150 mg/kg i.p.) showed substantial increases in NET protein expression. Maximal effect of vorinostat pretreatment in NB xenografts on 123I-MIBG biodistribution was observed in tumors that exhibited enhanced uptake in vorinostat-treated [0.062 ± 0.011 μCi/(mg tissue-dose injected)] vs. -untreated mice [0.022 ± 0.003 μCi/(mg tissue-dose injected); P < 0.05].

Conclusions: The results of our study provide preclinical evidence that vorinostat treatment can enhance NB therapy with 131I-MIBG. Clin Cancer Res; 17(8); 2339–49. ©2011 AACR.

Translational Relevance

In the preclinical results presented in this report, we validate the rationale of combining a histone deacetylase (HDAC) inhibitor, vorinostat, with the neuroblastoma targeted radiopharmaceutical agent, 131I-meta-iodobenzylguanidine (MIBG), for treatment of neuroblastoma (NB). Neuroblastomas are cancers of the sympathetic nervous system that generally retain expression of the norepinephrine transporter (NET). 131I-MIBG, a metabolically stable norepinephrine analog, is a substrate of NET and is in clinical trials for the treatment of NB. We demonstrate in in vitro and in vivo models that exposure to vorinostat causes increase in NET expression levels in NB tumors and enhanced uptake of 123I-MIBG. The vorinostat-induced increased 131I-MIBG uptake may result in enhanced efficacy of 131I-MIBG against NB. The results of this study provide strong support for coadministration of vorinostat and 131I-MIBG in the treatment of NB. A clinical trial for assessment of this combination in patients with resistant/relapsed neuroblastoma is already underway (NANT N2007-03).

Neuroblastoma (NB) is the most common extracranial solid tumor of childhood and is often diagnosed after metastasis has commenced (1). Originating from primitive sympathetic ganglion cells, NB can arise at any site in the sympathetic nervous system, with the adrenal gland being the most common site of origin. About 50% of all NB patients are classified as at “high risk,” with survival rates frequently being less than 40% with existing therapies (2). Patients with high-risk disease are at significant risk for resistance to initial therapy and have a high incidence of relapse subsequent to successful chemo- or radiotherapy (3). The unfavorable outcome for patients with relapsed or refractory disease creates an enormous need for novel therapeutic strategies with particularly high therapeutic indices for these patients.

As a sympathetic-derived tumor, NB cells typically possess capabilities of monoamine uptake, decarboxylation pathways for monoamine synthesis, and monoamine secretion (4). Of particular diagnostic and therapeutic value is the expression of the norepinephrine transporter (NET) (5). NET serves as an important uptake protein for the norepinephrine analog, meta-iodobenzylguanidine (MIBG), an agent used for the diagnosis and treatment of NB. The use of low-dose radiolabeled MIBG (typically 123I-MIBG) enables imaging to identify the sites and extent of neuroblastoma spread (6, 7). The use of high-dose radiolabeled MIBG (131I-MIBG) enables targeted radiotherapy for patients with neuroblastoma. Single agent studies have shown that 131I-MIBG is one of the most active agents for the treatment of patients with relapsed or refractory neuroblastoma (8).

The use of 131I-MIBG in NB is predicated on the concept that the radionuclide will selectively target NET expressing tumors and metastases and will not harm normal tissues, particularly those not expressing NET. A strategy built upon this concept would fundamentally carry the potential to deliver the high therapeutic index needed for successful NB treatment. The aims of the current study consisted of (i) mapping of NET in various tissues, (ii) chemical induction of NET expression, and (iii) study of 131I-MIBG uptake into tissues under the influence of a chemical NET expression modulator. The working hypothesis was that a chemical entity that results in enhanced expression of NET in NB cells would increase the concentration of 131I-MIBG in those sites and lead to preferential radiotherapy. Tissues that do not express NET could be expected to resist damage by 131I-MIBG.

One of the possible avenues of influencing the expression levels of NET was the inhibition of histone deacetylation by an agent like vorinostat (suberoylanilide hydroxamic acid, SAHA). Histone deacetylase (HDAC) inhibition in turn could be expected to cause generalized perturbation of cellular protein expression, possibly affecting NET expression. In this paper, we describe the results of proof-of-concept in vitro and in vivo experiments in human NB cell lines and NB xenografts. The results support our hypothesis, and have enabled us to obtain approval for the initiation of clinical trials that examine a combination therapy of vorinostat and 131I-MIBG for the treatment of neuroblastoma (NANT N2007-03).

Drugs and reagents

Radiolabeled 123I-MIBG was purchased from GE Healthcare. Radiolabeled 3H-norepinephrine was purchased from Perkin Elmer Life Sciences. All other chemicals were purchased from Sigma. Vorinostat was synthesized in our laboratory using a combination of previously published methods (9, 10). All the experiments described herein were also conducted utilizing a clinical grade sample of vorinostat obtained from Merck for comparison purposes. Antibodies for Western blotting were obtained from the following sources: rat polyclonal anti-NET antibody from Alpha Diagnostic International (NET11-A), horseradish peroxidase (HRP)-conjugated anti-rabbit (sc-2004; Santa Cruz Biotechnology) immunoglobulin was used as the secondary antibody and mouse monoclonal anti-β-actin antibody from Santa Cruz Biotechnology. Lipofectamine 2000 and hygromycin B were from Invitrogen. The cell culture media DMEM H-21, RPMI 1640, and FBS were obtained from the Cell Culture Facility of the University of California at San Francisco (San Francisco, CA).

Cell lines and transfection

Cell lines stably transfected with human NET, HEK-hNET, and empty vector, HEK-EV, were established by transfection of pcDNA5/FRT vector (Invitrogen) containing the full-length hNET cDNA, and pcDNA5/FRT vector alone, respectively, into human embryonic kidney 293 (HEK293) Flp-In cells using LipofectAMINE 2000 (Invitrogen) as per manufacturer's instructions. The stable clones were selected with 75 μg/mL hygromycin B. All the NB cell lines (LAN5, SHEP, SH-SY-5Y, Kelly, SK-N-SH, SK-N-MC, NB1691, and NB1691-luc) used in the present study were obtained from the UCSF Cell Culture Facility.

Cell culture

Stably transfected HEK293 cells were maintained in DMEM H-21 medium supplemented with 10% FBS, 100 units/mL penicillin and 100 units/mL streptomycin, and 75 μg/mL hygromycin B. The culture medium for all the NB cell lines was RPMI 1640 containing 15% FBS, 1% modified Eagle media (MEM) nonessential amino acids, 100 units/mL penicillin, and 100 μg/mL streptomycin. All cell lines were grown at 37°C in a humidified atmosphere with 5% CO2/95% air.

Cellular uptake of 3H-NE or 123I-MIBG

HEK-hNET cells were seeded in 24 well poly-d-lysine coated cell culture plates (BD Biosciences) in standard cell culture media and the uptake studies were conducted 24 hours after cell seeding. Cells were incubated in PBS buffer containing 15 nmol/L 3H-NE in the presence or absence of a NET specific inhibitor, desipramine (10 μmol/L), at 37°C for 3 minutes. After 3 minutes, uptake was terminated by rapid removal of the buffer containing the compounds and 3 quick washings with ice-cold PBS buffer. The cells were lysed with 1 mL of 0.1% SDS/0.1N NaOH solution for about 1 hour and the intracellular levels of radioactive NE were determined using liquid scintillation counting. Intracellular concentrations of norepinephrine were normalized to the total protein in each well using bicinchoninic acid (BCA) protein assay (Pierce).

The same procedure was followed for determination of 3H-NE or 123I-MIBG uptake rate in NB cell lines, SH-SY-5Y and Kelly. In this case, cells were exposed to increasing concentrations of vorinostat ranging from 0 to 5 μmol/L for 24 hours, followed by exposure to 3H-NE or 123I-MIBG in PBS buffer, at 37°C for 3 minutes. The intracellular levels of radioactivity corresponding to 3H-NE were determined as described earlier and were normalized to the total protein content in each well using BCA protein assay. The total concentration of 123I-MIBG was measured in an automated Wallac 1480 Wizard gamma counter using the program RiaCalc Wiz (Wallac Oy).

RNA isolation

Total RNA was isolated from each of the NB cell line on 6-well plates using the Invitrogen Micro-to-Midi Total RNA Purification System according to the manufacturer's protocol. Extracted RNA was stored at −80°C until use. Similarly, total RNA was isolated from 13 NB tumor samples. Samples of NB tumors were obtained from the UCSF Pediatric Solid Tumor Tissue Bank following procedures approved by the UCSF Committee on Human Research. Four samples were collected at initial diagnosis, 8 samples were collected after the initiation of therapy, and the timing of sample collection was unknown for 1 sample. Five of the tumor samples were from initial diagnosis (no prior treatment). All of the other samples were from patients who had received some type of therapy.

Reverse transcriptase PCR

Reverse transcriptase (RT) PCR of RNA samples was carried out with Superscript III (Invitrogen) using oligo(dT)20 primers. Two microliters of RT reaction product was used for subsequent PCR (Taq DNA Polymerase, Invitrogen) consisting of 35 cycles with the following parameters: 94°C for 30 seconds, 60°C for 45 seconds, 72°C for 1 minute, followed by a final extension of 72°C for 10 minutes and storage at 4°C. Primers were designed to amplify a unique sequence of human NET (SLC6A2), each spanning intron–exon boundaries to ensure no genomic DNA was amplified. The PCR primers that were used for human: NET (GenBank accession number NM_001043) forward: ATTCTTCAAAGGCGTTGGCTAT and reverse: CAGGACACCACGCTCATAAA were designed to amplify a 262-bp fragment. Analysis of each PCR sample was then performed on 2% agarose gels containing 0.5 μg/mL ethidium bromide. Gels were visualized using a digital camera and image processing system (Kodak). RT-PCR analysis for glyceraldehyde 3-phosphate dehydrogenase (GAPDH) gene was used as an internal control for each sample.

Quantitative real-time PCR

For quantitative real-time (qRT) PCR analysis, 2 μg of total RNA isolated from NB cells and tumor samples were reverse transcribed using High Capacity cDNA Reverse Transcription Kit (Applied Biosytems) in a 20-μL volume reaction according to the manufacturer's protocol. The resulting cDNA was used as template for qRT-PCR using TaqMan Gene Expression Assays for human transporter NET (Assay ID: Hs00426573_m1*) and human GAPDH (Assay ID: Hs99999905_m1). qRT-PCR reaction was carried out in 96-well reaction plates in a volume of 10 μL using the TaqMan Fast Universal Master Mix (Applied Biosystems). Reactions were run on the Applied Biosystems 7500 Fast Real-Time PCR System with the following profile: 95°C for 20 seconds followed by 40 cycles of 95°C for 3 seconds and 60°C for 30 seconds. The relative expression of each mRNA was calculated by the comparative method (ΔΔCt method). Firstly, the ΔCt value for each sample was obtained by subtracting the Ct value of GAPDH mRNA from the Ct value of the NET mRNA. Then the ΔΔCt value for each sample was obtained by subtracting the ΔCt value of another endogenous control, PGK1, from the ΔCt value of each sample. The mRNA expression level of NET was reported as the percentage of the expression of PGK1 calculated using the arithmetic formula 2−ΔΔCt (ABI PRISM 7700 Sequence Detection system User Bulletin No. 2, P/N 4303859) multiplied by 100.

Western blot analysis

The total protein extracts from transfected HEK293, NB cells and tumor samples were prepared using CellLytic M cell lysis buffer (Sigma) containing protease inhibitor cocktail at 4°C for 10 minutes. The cell homogenate was spun for 10 minutes at 14,000 rpm at 4°C and the protein concentration in the supernatant was determined by the BCA protein assay. Approximately 50 μg of the supernatant was resolved by SDS-PAGE and transferred onto polyvinylidene difluoride membrane (Bio-Rad). The blots were incubated for 1 hour in blocking buffer containing 5% nonfat dry milk in Tris-buffered saline (TBS) and then incubated overnight at 4°C with anti-human NET1 antibody diluted (1:750) in 2% milk in TBS buffer containing 0.1% Tween 20 (TBS-T). After washing with TBS-T, the blots were incubated for 1 hour at room temperature with the goat anti-rabbit IgG HRP-conjugated secondary antibody (1:2,500 dilution) in TBS-T buffer containing 5% milk. The blots were again washed thrice with TBS-T for 10 minutes and then developed with the ECL Western blotting detection reagent (GE Healthcare). All blots were reprobed for β-actin (1:10,000) as a loading control. Band intensities of scanned blots were quantified using ImageJ. The integrated intensity of a fixed area was measured, and background levels were subtracted.

Cytotoxicity studies

The cytotoxicity of MIBG was measured by standard MTT assays in 96-well plates using NB cell lines. After seeding the cells at the desired density and incubating overnight, vehicle control [5% DMSO (dimethyl sulfoxide) in PBS] or vorinostat (5 μmol/L) was added to the culture medium. After 24 hours of treatment, the drug-containing medium was replaced with medium containing MIBG at various concentrations and the incubation was continued for additional 72 hours. At the end of the incubation, 20 μL of MTT stock solution (5 mg/mL) was added to each well. After additional incubation for 3 hours at 37°C, the MTT reaction medium was discarded and the purple-blue MTT formazan crystals were dissolved by the addition of 100 μL of 0.1N HCI in isopropanol. The optical density, which is a measure of the mitochondrial function of the viable cells, was read directly with a microplate reader (model versamax; Molecular Devices Co.) at 580 nm and a reference wavelength of 680 nm. Concentration response graphs were generated for each drug using GraphPad Prism software (GraphPad Software, Inc.). These graphs were analyzed using a curve fit for sigmoid dose–response, and IC50 values were derived. Results are expressed as mean IC50 values with the standard error of the mean.

Animals

Female CrTac:NCr-Foxn1nu athymic mice (4–5 weeks old) were purchased from Taconic and housed under aseptic conditions, which included filtered air, sterilized food, water, bedding, and cages. The experiments on mice were approved by the Institutional Animal Care and Use Committee of University of California at San Francisco. All experimental and animal handling procedures were in accordance with national ethical guidelines.

Preparation of neuroblastoma xenografts and drug treatment

To study the effect of vorinostat treatment on NET expression in vivo, nude mice harboring xenografts of human NB1691 neuroblastoma cells expressing luciferase (NB1691-luc) were used. For preparation of NB xenografts, NB1691-luc cells were resuspended in serum free RPMI1640 media and 100 μL of this mixture containing 6 × 106 cells were injected in mice by intravenous tail vein injection. Serial tumor measurements were obtained by weekly bioluminescence imaging after an intraperitoneal injection of d-luciferin (Xenogen) at 15 mg/mL in sterile PBS. Twenty-one days after injection of tumor cells, the animals were divided into vehicle control (5% DMSO in saline) and vorinostat-treated (150 mg/kg i.p.) groups (2 per group). Tumors were then harvested from each animal at various time points (1, 3, 6, and 18 hours) after initiation of treatment and lysed with a buffer containing (20 mmol/L Tris–HCl pH 7.5–8.0, 150 mmol/L NaCl, 0.5% sodium deoxycolate, 1% Triton X-100, 0.1 SDS, 1 mmol/L EDTA, 1 mmol/L phenylmethylsulphonyl fluoride, and protease inhibitor cocktail). qRT-PCR and Western blot analysis of these tissues was carried out as described under the Quantitative real-time PCR and Western blot analysis sections.

In order to study the effect of vorinostat administration on tissue expression of NET in healthy mice not bearing tumors, nude mice (5 per group) were injected with vorinostat (150 mg/kg i.p.) and various tissues were harvested 6 and 18 hours after drug treatment. Each of these tissues was then subjected to total RNA extraction and qRT-PCR analysis as described before.

Biodistribution measurements of 123I-MIBG

Animals with NB xenografts (described above) were divided into 2 groups (3 per group) in the following manner: (i) control group: injected with vehicle (5% DMSO in saline i.p.) 6 hours prior to 123I-MIBG (300 μCi/mouse) by intravenous tail vein injection; (ii) treatment group: injected with vorinostat (150 mg/kg i.p.) 6 hours before intravenous tail vein injection of 123I-MIBG. Two hours after the treatment, the mice were euthanized. Samples of liver, kidney, spleen, heart, and tumor were excised, weighed, and their associated radioactivity was measured in an automated Wallac 1480 Wizard gamma counter using the program RiaCalc Wiz (Wallac Oy). Blood was also sampled and radioactivity content was normalized to its weight. The uptake of 123I-MIBG was expressed as the fraction of the injected dose per gram of tissue. A correction was made for radioactive decay, which would take place since the time of injection.

Data analysis

Data were analyzed statistically by unpaired or paired Student t tests, as appropriate. Probability values lower than 0.05 were considered statistically significant.

123I-MIBG uptake was increased in hNET transfected cells

Being an analog of monoamine neurotransmitter norepinephrine, the transport of MIBG by NET is well documented (11). We confirmed the uptake of MIBG by NET in our in vitro model using HEK293 cells stably transfected with human NET transporter (HEK-hNET). Due to its ease of handling and shorter half life, we used 123I-labeled MIBG instead of 131I-labeled MIBG in our in vitro and in vivo studies (12). The cellular accumulation of 123I-MIBG in HEK-hNET cells [0.484 ± 0.033 μCi/(mg protein min)] was 13.4-fold higher than in HEK-EV cells [0.036 ± 0.003 μCi/(mg protein min); P < 0.0001; Supporting Information; Supplementary Fig. S1]. Incubation of 123I-MIBG with a NET inhibitor (desipramine, 10 μmol/L) significantly reduced the cellular accumulation of MIBG in hNET expressing cells [0.042 ± 0.0033 μCi/(mg protein min); P < 0.0001], with no effect in HEK-EV cells [0.030 ± 0.0026 μCi/(mg protein min); P > 0.05].

Neuroblastoma cell lines and tumor samples show high NET expression

Originating from the sympathetic nervous system, neuroblastomas retain the ability of neuronal cells to uptake NE through the neuronal NE transporter. Hence the baseline expression level of NET transcripts in 7 NB cell lines and 13 NB tumor samples was examined by qRT-PCR analysis. All the NB cell lines tested showed detectable and variable expression of NET mRNA. The highest expression of NET was observed in LAN5, SK-N-SH, and NB1691 cell lines and was in the range of 23% to 45% of the expression of PGK1 (3-phosphoglycerate kinase), an abundantly expressed endogenous control (Fig. 1A). NET mRNA was detected in 12 out of 13 NB tumor samples across a range of expression levels. NET transcripts were expressed at more than 40% of the endogenous control PGK1 in 9 out of the 13 tumor samples (Fig. 1B). The comparison of NET transcripts with the endogenous control signifies the high abundance and significance of NET protein to NB cell types.

Figure 1.

mRNA expression level of the NET, in neuroblastoma cell lines (A) and tumor samples (B). Total RNA was isolated from 7 neuroblastoma cell lines and 13 neuroblastoma patient tumor samples. The NET mRNA expression was quantified by qRT-PCR as described in the Materials and Methods section. The data presented is from a single experiment and is reported as the percentage of expression of PGK1, an endogenous control. Similar results were obtained in 3 independent experiments.

Figure 1.

mRNA expression level of the NET, in neuroblastoma cell lines (A) and tumor samples (B). Total RNA was isolated from 7 neuroblastoma cell lines and 13 neuroblastoma patient tumor samples. The NET mRNA expression was quantified by qRT-PCR as described in the Materials and Methods section. The data presented is from a single experiment and is reported as the percentage of expression of PGK1, an endogenous control. Similar results were obtained in 3 independent experiments.

Close modal

Vorinostat causes a dose-dependent increase in the levels of expression of NET mRNA and protein in neuroblastoma cell lines

To determine the effect of vorinostat treatment on the expression of NET, we cultured 2 NB cell lines in the presence of either DMSO (vehicle control) or vorinostat (0.05, 0.10, 0.25, 0.50, 0.75, 1.00, 2.5, and 5.00 μmol/L) for 24 hours. The cell lines selected for this purpose (SH-SY-5Y and Kelly) were based on their moderate NET expression and to allow evaluation of MYCN amplified (Kelly) and MYCN nonamplified (SH-SY-5Y) status of these cell lines. The doses of vorinostat were based on clinically relevant plasma concentrations (13, 14). RT-PCR analysis of mRNA isolated from vorinostat-treated cells demonstrated qualitatively a dose-dependent increase in the expression of NET transcripts in both of the cell lines (Fig. 2A).

Figure 2.

Enhanced expression levels of NET mRNA in neuroblastoma cell lines treated with vorinostat by RT-PCR (A) and qRT-PCR (B). SH-SY-5Y and Kelly neuroblastoma cells were treated with increasing concentrations of vorinostat ranging from 0 to 5 μmol/L for 24 hours, followed by RNA purification and RT-PCR or qRT-PCR using NET-specific primers as described in the Materials and Methods section. Amplification of GAPDH was used as a loading control for RT-PCR. C, enhanced expression of NET protein in neuroblastoma cell lines exposed to vorinostat. Cell extracts from Kelly and SH-SY-5Y cells treated and untreated with vorinostat (1 and 5 μmol/L) for 24 hours were prepared and 50 μg of protein lysate from each sample was evaluated by Western blot as described in the Materials and Methods section. β-Actin was used as a loading control.

Figure 2.

Enhanced expression levels of NET mRNA in neuroblastoma cell lines treated with vorinostat by RT-PCR (A) and qRT-PCR (B). SH-SY-5Y and Kelly neuroblastoma cells were treated with increasing concentrations of vorinostat ranging from 0 to 5 μmol/L for 24 hours, followed by RNA purification and RT-PCR or qRT-PCR using NET-specific primers as described in the Materials and Methods section. Amplification of GAPDH was used as a loading control for RT-PCR. C, enhanced expression of NET protein in neuroblastoma cell lines exposed to vorinostat. Cell extracts from Kelly and SH-SY-5Y cells treated and untreated with vorinostat (1 and 5 μmol/L) for 24 hours were prepared and 50 μg of protein lysate from each sample was evaluated by Western blot as described in the Materials and Methods section. β-Actin was used as a loading control.

Close modal

Using qRT-PCR analysis the increase in NET expression after vorinostat treatment observed earlier was quantified (Fig. 2B). Even the lowest concentration of vorinostat (0.025 μmol/L) resulted in a 2-fold increase in NET expression in Kelly cells over the nontreated cells and a maximum increase of 6.7-fold was achieved at 5 μmol/L dose of vorinostat. A dose-dependent increase in NET mRNA expression was also observed in SH-SY-5Y cells after vorinostat treatment with maximum attainable increase of 5.6-fold after exposure to 5 μmol/L concentration of vorinostat. The increase in the expression of NET mRNA observed by RT-PCR and qRT-PCR data, however, does not necessarily prove the presence of functional NET transporter protein. These studies were therefore supplemented with Western blotting and functional characterization of elevated NET protein after vorinostat treatment in NB cell lines.

Increases in NET mRNA expression in NB cells after vorinostat treatment were examined for correlations with NET protein levels. NET protein concentrations in Kelly and SH-SY-5Y cells exposed to 1 and 5 μmol/L vorinostat for 24 hours were analyzed by Western blot (Fig. 2C). The increase in NET mRNA levels observed in both the NB cell lines was in agreement with the increase in NET protein synthesis. Kelly and SH-SY-5Y cells treated with vorinostat (5 μmol/L) showed approximately 5.3- and 6.9-fold increases in NET protein compared to untreated cells, respectively.

Vorinostat treatment increases uptake of NET substrates in vitro

We next performed a functional characterization of NET following vorinostat treatment. The cellular accumulation of a protypical NET substrate, norepinephrine (NE), was determined in the presence and absence of a range of vorinostat concentrations in 2 NB cell lines (Fig. 3A). The cellular accumulation rate of 3H-NE (15 nmol/L) in NB cells treated for 24 hours at the highest vorinostat concentration (5 μmol/L) was 2.5- to 3.4-fold higher than the corresponding untreated control cells. Specifically, the uptake of 3H-NE in Kelly cells in the presence and absence of vorinostat (5 μmol/L) treatment was 0.252 ± 0.017 nmol/(mg protein.min) and 0.009 ± 0.006 nmol/(mg protein.min), respectively (P < 0.001). Similarly, the highest concentration of vorinostat resulted in an accumulation rate of 0.178 ± 0.011 nmol/(mg protein.min) for 3H-NE in SH-SY-5Y cells, which was significantly greater than 0.052 ± 0.004 nmol/(mg protein.min) in untreated cells (P < 0.001). Both cell lines exhibited a dose-dependent increase in NE uptake across the range of evaluated vorinostat concentrations (0–5 μmol/L).

Figure 3.

Increased uptake of 3H-norepinephrine (A) and 123I-MIBG (B) in neuroblastoma cell lines after exposure to vorinostat. Kelly (black bars) and SH-SY-5Y (white bars) cells were exposed to increasing concentrations of vorinostat ranging from 0 to 5 μmol/L for 24 hours, followed by uptake of 3H-NE or 123I-MIBG for 3 minutes. The net uptake is represented as fold over control, where control is the uptake in neuroblastoma cells in the absence of vorinostat pretreatment. Data are expressed as the mean ± SEM from 3 independent experiments.

Figure 3.

Increased uptake of 3H-norepinephrine (A) and 123I-MIBG (B) in neuroblastoma cell lines after exposure to vorinostat. Kelly (black bars) and SH-SY-5Y (white bars) cells were exposed to increasing concentrations of vorinostat ranging from 0 to 5 μmol/L for 24 hours, followed by uptake of 3H-NE or 123I-MIBG for 3 minutes. The net uptake is represented as fold over control, where control is the uptake in neuroblastoma cells in the absence of vorinostat pretreatment. Data are expressed as the mean ± SEM from 3 independent experiments.

Close modal

Similarly, vorinostat pretreatment (0–5 μmol/L) caused a dose-dependent increase in 123I-MIBG uptake in the NB cells. In the absence of vorinostat treatment, the intracellular concentration of 123I-MIBG after its exposure for 3 minutes to Kelly cells was 0.025 ± 0.001 μCi/(mg protein.min). This intracellular concentration of 123I-MIBG increased 3.9-fold after pretreatment with 5 μmol/L vorinostat [0.097 ± 0.018 μCi/(mg protein.min); P < 0.001]. Similarly, a 2.6-fold enhancement in 123I-MIBG uptake was observed in SH-SY-5Y cells pretreated with vorinostat [5 μmol/L; 0.062 ± 0.003 μCi/(mg protein.min)] compared to untreated cells [0.024 ± 0.001 μCi/(mg protein.min); P < 0.001; Fig. 3B]. Furthermore, the increase in cellular uptake of NE and MIBG observed after vorinostat pretreatment was inhibited by desipramine (10 μmol/L), a NET inhibitor (data not shown). When these uptake experiments were performed under various preincubation periods or vorinostat (2, 4, 6, 12, and 24 hours), the maximum changes in the uptake were observed at the 6-hour time point with a negligible further increase at 24 hours (Supporting Information; Supplementary Fig. S4), in agreement with maximum increase in NET mRNA and protein observed at that time point (Supporting Information; Supplementary Fig. S3). The vorinostat pretreatment period of 24 hours was selected to capture maximal changes in the uptake of NET substrates and for simplicity of experimentation.

Cytotoxic efficacy of MIBG is enhanced in the presence of vorinostat

To obtain a correlation between vorinostat-induced increases in MIBG uptake in NB cell lines via NET upregulation and the functional consequences of this enhanced uptake on cell growth, we examined the cytotoxicity of MIBG in the presence and absence of vorinostat. The results of standard MTT assays indicate that MIBG in combination with vorinostat is prominently more toxic against all NB cell lines tested when compared to MIBG alone (Table 1). The IC50 value of MIBG in the presence of vorinostat is 1.88 to 3.57 (depending on the NB cell line) order of magnitude lower than its IC50 as a single agent. Changes in cell proliferation following exposure to 5 μmol/L vorinostat were less than 10% for 36-hour exposure periods and less than 20% for 48 hours.

Table 1.

Enhanced efficacy of MIBG in the presence of vorinostat

Cell lineIC50, μmol/L
MIBGMIBG + vorinostatResistance factor
SHEP 70.8 ± 4.51 26.4 ± 2.31 2.68a 
Kelly 58.5 ± 3.54 18.3 ± 1.93 3.20a 
SH-SY-5Y 52.2 ± 2.24 14.6 ± 1.39 3.57b 
SK-N-SH 35.1 ± 5.58 17.4 ± 1.72 2.02c 
NB1691 43.0 ± 5.94 22.9 ± 2.99 1.88c 
Cell lineIC50, μmol/L
MIBGMIBG + vorinostatResistance factor
SHEP 70.8 ± 4.51 26.4 ± 2.31 2.68a 
Kelly 58.5 ± 3.54 18.3 ± 1.93 3.20a 
SH-SY-5Y 52.2 ± 2.24 14.6 ± 1.39 3.57b 
SK-N-SH 35.1 ± 5.58 17.4 ± 1.72 2.02c 
NB1691 43.0 ± 5.94 22.9 ± 2.99 1.88c 

NOTE: NB cell lines were pretreated with or without vorinostat (5 μmol/L) for 24 hours before treatment with MIBG for 3 days. Decrease in the IC50 of MIBG was observed when vorinostat was present. The enhancement in MIBG efficacy is represented as the resistance factor which is the ratio of IC50 of MIBG in the absence and presence of vorinostat. The data are represented as the mean ± SEM of 3 independent experiments.

aP < 0.001.

bP < 0.0001.

cP < 0.05.

Vorinostat treatment caused increases in NET protein levels in neuroblastoma xenografts in mice

The next aim for this study was to obtain in vivo proof-of-principle of the observed vorinostat-induced NET upregulation and its functional consequences. Neuroblastoma xenografts created by intravenous tail vein injection of NB1691-luc cells were used for these experiments. After allowing tumors to develop for 21 days, mice were injected with 150 mg/kg of vorinostat or vehicle control intraperitoneally. Tumors harvested at various time points after vorinostat administration were analyzed by Western blotting (Fig. 4A). A 2.1- and 2.5-fold increase in NET protein level compared to the untreated control mice was noticeable at the 1- and 3-hour time points, respectively, in the vorinostat-treated mice. The tumors at 6- and 18-hour time points exhibited 3.8- and 4.2-fold increases in NET protein with vorinostat treatment over the untreated control mice, respectively (Supporting Information; Supplementary Fig. S5A). Similarly, the corresponding changes in NET mRNA expression levels were observed in these samples (Supporting Information; Supplementary Fig. S5B). The rapid tumor progression observed in this metastatic mouse model and negligible differences in NET expression enhancement prompted us to use the 6-hour time point for biodistribution studies. No change in NET protein expression level was observed in mice treated with the vehicle control over the entire period of the experiment.

Figure 4.

A, enhanced expression of NET protein in neuroblastoma tumors treated with vorinostat in vivo. Mice were injected with NB1691-luc cells and after significant tumor development, mice were divided into vehicle control (5% DMSO in saline) and vorinostat-treated (150 mg/kg i.p.) groups. Tumors were harvested at 1, 3, 6, and 18 hours after vorinostat treatment and expression of NET protein was determined by Western blot analysis as described in the Materials and Methods section. B, enhanced expression of NET in tissues of healthy Foxn1nu mice 6 and 18 hours after intraperitoneal injection of vorinostat (150 mg/kg). Total RNA was isolated from each tissue and quantified for NET mRNA level by qRT-PCR as described in the Materials and Methods section. Data are represented as the mean ± SEM.

Figure 4.

A, enhanced expression of NET protein in neuroblastoma tumors treated with vorinostat in vivo. Mice were injected with NB1691-luc cells and after significant tumor development, mice were divided into vehicle control (5% DMSO in saline) and vorinostat-treated (150 mg/kg i.p.) groups. Tumors were harvested at 1, 3, 6, and 18 hours after vorinostat treatment and expression of NET protein was determined by Western blot analysis as described in the Materials and Methods section. B, enhanced expression of NET in tissues of healthy Foxn1nu mice 6 and 18 hours after intraperitoneal injection of vorinostat (150 mg/kg). Total RNA was isolated from each tissue and quantified for NET mRNA level by qRT-PCR as described in the Materials and Methods section. Data are represented as the mean ± SEM.

Close modal

The effect of vorinostat on NET transcription was studied in different tissues of healthy mice at 6 and 18 hours following intraperitoneal vorinostat administration (Fig. 4B). The greatest relative increases in NET mRNA expression levels were in the brain (7.82-fold over untreated control) and in the liver (7.69-fold over untreated control) after 6 hours. At the 18-hour time point, increases in NET mRNA levels were also observed in the adrenal gland and skin (3.64- and 2.25-fold over untreated controls, respectively).

Biodistribution of 123I-MIBG

The functional effect of increased NET protein level after vorinostat treatment was next evaluated in terms of changes in MIBG in vivo biodistribution. Figure 5 presents the biodistribution of 123I-MIBG 2 hours after intravenous administration in nude mice bearing NB1691-luc human neuroblastoma xenografts. Half of these xenografts received pretreatment with intraperitoneal vorinostat (150 mg/kg) 6 hours before MIBG administration. Differences in tissue accumulation due to vorinostat administration reaching statistical significance were observed only in the liver and in tumors. Without vorinostat treatment, the concentration of MIBG in the liver was 0.025 ± 0.003 μCi/(mg tissue-dose injected) whereas with vorinostat pretreatment, the total concentration of MIBG in the liver increased to 0.049 ± 0.006 μCi/(mg tissue-dose injected) (P < 0.05; Fig. 5). The greatest increases in MIBG accumulation due to vorinostat treatment were observed in tumors, which were 2.8-fold higher than the corresponding control group [with vorinostat pretreatment: 0.062 ± 0.011 μCi/(mg tissue-dose injected) vs. untreated: 0.022 ± 0.003 μCi/(mg tissue-dose injected); P < 0.05; Fig. 5]. Although there was a trend toward increase in MIBG concentration after vorinostat pretreatment in other tissues (e.g., kidney, spleen, heart), the difference in concentrations between the vorinostat-treated and -untreated group did not reach statistical significance. A trend was also observed toward reduced MIBG accumulation in the blood in the vorinostat pretreated group, but the difference did not reach statistical significance [with vorinostat pretreatment: 0.004 ± 0.001 μCi/(mg tissue-dose injected) vs. no vorinostat pretreatment: 0.007 ± 0.001 μCi/(mg tissue-dose injected); P > 0.05; Fig. 5]. Because the compound does not cross the BBB, MIBG brain accumulation was below the detection limit of the assay (15).

Figure 5.

Biodistribution of 123I-MIBG in Foxn1nu mice after vorinostat treatment. Neuroblastoma xenografts generated by intravenous tail vein injection of NB1691-luc cells in mice were treated with vorinostat (150 mg/kg i.p.; black bars) and vehicle control (5% DMSO in saline; white bars). After 6 hours, 123I-MIBG (300 μCi/mouse) was injected by intravenous tail vein and tissues were collected after 2 hours to measure the accumulation of MIBG as described in the Materials and Methods section. Data are represented as the mean ± SEM (*, P < 0.05).

Figure 5.

Biodistribution of 123I-MIBG in Foxn1nu mice after vorinostat treatment. Neuroblastoma xenografts generated by intravenous tail vein injection of NB1691-luc cells in mice were treated with vorinostat (150 mg/kg i.p.; black bars) and vehicle control (5% DMSO in saline; white bars). After 6 hours, 123I-MIBG (300 μCi/mouse) was injected by intravenous tail vein and tissues were collected after 2 hours to measure the accumulation of MIBG as described in the Materials and Methods section. Data are represented as the mean ± SEM (*, P < 0.05).

Close modal

We have demonstrated that exposure to vorinostat at clinically relevant concentrations enhances NET mRNA and protein expression in neuroblastoma cell lines and tumor xenografts. This enhanced NET expression translates into increases in MIBG uptake into model systems, suggesting that the newly synthesized NET protein is functional. While increased MIBG uptake was observed in nontumor tissues, the greatest vorinostat-induced increase in uptake was in neuroblastoma tumors. This suggests a possible favorable therapeutic index for 131I-MIBG when administered together with vorinostat. We hypothesize that combination of vorinostat and 131I-MIBG will augment intracellular accumulation of 131I-MIBG in patients with neuroblastoma. This hypothesis forms the basis for an ongoing phase 1 clinical trial of this combination in children and young adults with relapsed or refractory neuroblastoma (NANT N2007-03).

Results from our laboratory and others clearly demonstrate that NET is expressed at higher than endogenous levels in almost all NB tumors evaluated (Fig. 1). MIBG, a metabolically stable NE analog, accumulates in neuroendocrine tissues via NET (Supporting Information; Supplementary Fig. S1) and MIBG labeled with either 131I or 123I is used for the scintigraphic visualization (diagnosis) and therapy of pheochromocytomas and neuroblastomas (16–18). The polar guanidinium group borne by MIBG precludes its movement across the BBB into the brain (15), making MIBG highly appropriate for our aim of achieving a high therapeutic index.

The experiments in this study were designed to evaluate the hypothesis that chemically induced upregulation of NET would enhance partitioning of MIBG into NB tumors. The chemical induction of NET expression was inhibition of HDAC through administration of vorinostat. RT-PCR, qRT-PCR, and Western blot analysis of NB cells treated with increasing concentrations of vorinostat revealed dose-dependent increases in NET transcription and translation (Fig. 2). The mechanism through which vorinostat upregulates NET expression remains unclear. Vorinostat chelates an essential Zn2+ ion, deactivating a host of HDAC isoenzymes. HDAC inhibition causes chromatin opening, making it more accessible to transcription factors and thereby leading to increased expression of cellular proteins. However, due to the intricate secondary interrelationships between cellular transcription factors and cellular proteins, the net result may not necessarily be upregulation of a particular protein. For example, HDAC inhibition may also promote increased acetylation of transcription factors, thereby increasing their activity, e.g., in the case of the tumor suppressor p53, HDAC inhibition increases its activity, which in turn may lead to cell cycle arrest and even apoptosis (19). The pathway through which vorinostat upregulates NET may therefore involve several levels of interplay between transcription factors and chromatin opening. Vorinostat is already approved for the treatment of cutaneous T cell lymphoma and has shown moderate activity as a single agent against neuroblastoma cell lines in preclinical studies (20). Furthermore a large body of preclinical data also indicates that HDAC inhibitors including vorinostat can act as radiosensitizers across a range of human cancers (21–24). Combinations of 131I-MIBG with radiosensitizing agents like topotecan (25) and irinotecan (NANT2004-06) have been successfully evaluated and shown to be tolerable in clinical trials. These results prompted us to evaluate vorinostat/131I-MIBG combination for NB. Vorinostat pretreatment indeed caused increased accumulation of MIBG in NB cells (Fig. 3). The cytotoxicity of MIBG also was vastly enhanced when NB cells were pretreated with vorinostat as opposed to MIBG alone (Table 1). These data conclusively link the increased cellular accumulation of MIBG in the presence of vorinostat with a corresponding increase in its toxicity.

NB tumors are biologically heterogeneous, and may in theory respond dissimilarly to vorinostat exposure. A clinically utilized indicator of NB prognosis is MYCN gene amplification and is an indication of advanced disease (26, 27). In order to address some of this heterogeneity, we evaluated 2 cell lines that differ in their MYCN status: the Kelly cell line is MYCN amplified while the SH-SY-5Y cell line is not. Throughout the experiments, vorinostat-induced NET upregulation was independent of the MYCN status of the cell line. It should, however, be noted that HDAC inhibitors such as valproic acid have been previously shown to downregulate MYCN (28). Indeed, vorinostat was found to downregulate MYCN (data not shown) suggesting that any underlying correlation between NET upregulation and MYCN status may be masked by the effect of vorinostat on MYCN.

The mapping of vorinostat-induced NET overexpression in vivo demonstrated increase in NET mRNA transcription in the brain, liver, adrenal gland, and skin for a healthy mouse (Fig 4B). This effect was prominent even 6 hours after vorinostat administration. Similar increases in NET protein levels was observed in NB tumors at 6-hour time-point (Fig. 4A) which correlated with the significantly increased MIBG uptake in the tumors of vorinostat-treated mice over untreated controls (Fig. 5). Although MIBG uptake also increased in the livers of vorinostat-treated mice, the animal model used in our study contained high degrees of tumor metastasis. The increased MIBG uptake in the liver may possibly have occurred due to the contribution of NB cells metastatic to the liver. 131I-MIBG is associated with a very low-incidence of hepatic toxicity (29). The current findings nevertheless raise the possibility of increased hepatic toxicity when 131I-MIBG is combined with vorinostat. Clinical trials of the combination will need to monitor for this possibility.

The main dose limiting toxicity of 131I-MIBG is hematologic toxicity. We observed a trend toward decreased plasma concentrations of MIBG in mice treated with vorinostat (Fig. 5). While we were not able to evaluate bone marrow levels of MIBG, it is possible that the combination of vorinostat and 131I-MIBG may result in less hematologic toxicity through 2 mechanisms. First, if the combination results in increased 131I-MIBG into tumor cells, then a lower dose of 131I-MIBG may be as efficacious as 131I-MIBG given as a single agent at a higher dose. In addition, if bone marrow uptake of 131I-MIBG is reduced by vorinostat treatment though preferential channeling of 131I-MIBG into tumors, then hematologic toxicity may also be reduced.

In spite of many combination therapies tried in the treatment of neuroblastoma, the goal of a very high therapeutic index with sparing of the normal and the developing tissues remains elusive. The strategy designed and developed in this study is perhaps one of the few that are derived specifically from addressing each of the unique aspects of neuroblastoma that differentiate them from other tumors. While the enhancement of MIBG uptake by vorinostat comes about from a biochemical change induced by vorinostat, the sparing of CNS results from biodistribution aspects of MIBG. Through selective partitioning of MIBG into NB tumors by vorinostat administration, it may be possible to achieve desired efficacy from a lower dose of MIBG, potentially mitigating the problematic hematologic toxicity often observed with MIBG therapy.

No potential conflicts of interest were disclosed.

This work was supported by NIH grants GM61390, GM36780, PO1 81403 (K.K. Matthay and W.A. Weiss), R01CA102321 (W.A. Weiss) and KL2 RR024130; by a grant (17RT-0126) from the Tobacco-Related Disease Research Program (TRDRP); and by the Alex Lemonade, Dougherty and the Campini Foundation.

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.
Goodman
MT
,
Gurney
JG
,
Smith
MA
,
Olshan
AF
. 
Sympathetic nervous system tumors
.
In:
Ries
LA
,
Smith
MA
,
Gurney
JG
, et al
editors
. 
Cancer Incidence and Survival among Children and Adolescents: United States SEER Program, 1975–1995
.
Bethesda, MD
:
National Cancer Institute
; 
1999
.
p. 65
72
.
2.
Maris
JM
. 
Recent advances in neuroblastoma
.
N Engl J Med
2010
;
362
:
2202
11
.
3.
DuBois
SG
,
Kalika
Y
,
Lukens
JN
,
Brodeur
GM
,
Seeger
RC
,
Atkinson
JB
, et al
Metastatic sites in stage IV and IVS neuroblastoma correlate with age, tumor biology, and survival
.
J Pediatr Hematol Oncol
1999
;
21
:
181
9
.
4.
Takeda
N
. 
Simultaneous analyses of monoamines and their metabolites in urine specimens of patients with neuroblastoma
.
Comp Biochem Physiol C
1993
;
105
:
373
7
.
5.
Hadrich
D
,
Berthold
F
,
Steckhan
E
,
Bönisch
H
. 
Synthesis and characterization of fluorescent ligands for the norepinephrine transporter: potential neuroblastoma imaging agents
.
J Med Chem
1999
;
42
:
3101
8
.
6.
Taggart
D
,
Dubois
S
,
Matthay
KK
. 
Radiolabeled metaiodobenzylguanidine for imaging and therapy of neuroblastoma
.
Q J Nucl Med Mol Imaging
2008
;
52
:
403
18
.
7.
DuBois
SG
,
Matthay
KK
. 
Radiolabeled metaiodobenzylguanidine for the treatment of neuroblastoma
.
Nucl Med Biol
2008
;
35
(
Suppl. 1
):
S35
48
.
8.
Matthay
KK
,
Yanik
G
,
Messina
J
,
Quach
A
,
Huberty
J
,
Cheng
SC
, et al
Phase II study on the effect of disease sites, age, and prior therapy on response to iodine-131-metaiodobenzylguanidine therapy in refractory neuroblastoma
.
J Clin Oncol
2007
;
25
:
1054
60
.
9.
Gediya
LK
,
Chopra
P
,
Purushottamachar
P
,
Maheshwari
N
,
Njar
VC
. 
A new simple and high-yield synthesis of suberoylanilide hydroxamic acid and its inhibitory effect alone or in combination with retinoids on proliferation of human prostate cancer cells
.
J Med Chem
2005
;
48
:
5047
51
.
10.
Vartak
AP
,
Skoblenick
K
,
Thomas
N
,
Mishra
RK
,
Johnson
RL
. 
Allosteric modulation of the dopamine receptor by conformationally constrained type VI beta-turn peptidomimetics of Pro-Leu-Gly-NH2
.
J Med Chem
2007
;
50
:
6725
9
.
11.
Bayer
M
,
Kuçi
Z
,
Schömig
E
,
Gründemann
D
,
Dittmann
H
,
Handgretinger
R
, et al
Uptake of MIBG and catecholamines in noradrenaline- and organic cation transporter-expressing cells: potential use of corticosterone for a preferred uptake in neuroblastoma- and pheochromocytoma cells
.
Nucl Med Biol
2009
;
36
:
287
94
.
12.
Mandel
SJ
,
Shankar
LK
,
Benard
F
,
Yamamoto
A
,
Alavi
A
. 
Superiority of iodine-123 compared with iodine-131 scanning for thyroid remnants in patients with differentiated thyroid cancer
.
Clin Nucl Med
2001
;
26
:
6
9
.
13.
Fouladi
M
,
Park
JR
,
Stewart
CF
,
Gilbertson
RJ
,
Schaiquevich
P
,
Sun
J
, et al
Pediatric phase I trial and pharmacokinetic study of vorinostat: a Children's Oncology Group phase I consortium report
.
J Clin Oncol
2010
;
28
:
3623
9
.
14.
Kelly
WK
,
O'Connor
OA
,
Krug
LM
,
Chiao
JH
,
Heaney
M
,
Curley
T
, 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
.
15.
Gourand
F
,
Mercey
G
,
Ibazizène
M
,
Tirel
O
,
Henry
J
,
Levacher
V
, et al
Chemical delivery system of metaiodobenzylguanidine (MIBG) to the central nervous system
.
J Med Chem
2010
;
53
:
1281
7
16.
Shapiro
B
,
Gross
MD
. 
Radiochemistry, biochemistry, and kinetics of 131I-metaiodobenzylguanidine (MIBG) and 123I-MIBG: Clinical implications of the use of 123I-MIBG
.
Med Ped Oncol
1987
;
15
:
170
7
.
17.
Hoefnagel
CA
,
Voute
PA
,
DeKraker
J
,
Marcuse
HR
. 
Radionuclide diagnosis and therapy of neural crest tumors using iodine-131 metaiodobenzylguanidine
.
J Nucl Med
1987
;
28
:
308
14
.
18.
Klingenbiel
T
,
Bader
P
,
Bares
R
,
Beck
J
,
Hero
B
,
Jürgens
H
, et al
Treatment of neuroblastoma stage 4 with 131I-meta-iodo-benzylguanidine, high-dose chemotherapy and immunotherapy. A pilot study
.
Eur J Cancer
1998
;
34
:
1398
402
.
19.
Harms
KL
,
Chen
X
. 
Histone deacetylase 2 modulates p53 transcriptional activities through regulation of p53-DNA binding activity
.
Cancer Res
2007
;
67
:
3145
52
.
20.
De los Santos
M
,
Zambrano
A
,
Aranda
A
. 
Combined effects of retinoic acid and histone deacetylase inhibitors on human neuroblastoma SH-SY5Y cells
.
Mol Cancer Ther
2007
;
6
:
1425
32
.
21.
Camphausen
K
,
Tofilon
PJ
. 
Inhibition of histone deacetylation: a strategy for tumor radiosensitization
.
J Clin Oncol
2007
;
25
:
4051
6
.
22.
Chinnaiyan
P
,
Vallabhaneni
G
,
Armstrong
E
,
Huang
SM
,
Harari
PM
. 
Modulation of radiation response by histone deacetylase inhibition
.
Int J Radiat Oncol Biol Phys
2005
;
62
:
223
9
.
23.
Flatmark
K
,
Nome
RV
,
Folkvord
S
,
Bratland
A
,
Rasmussen
H
,
Ellefsen
MS
, et al
Radiosensitization of colorectal carcinoma cell lines by histone deacetylase inhibition
.
Radiat Oncol
2006
;
1
:
25
.
24.
Munshi
A
,
Tanaka
T
,
Hobbs
ML
,
Tucker
SL
,
Richon
VM
,
Meyn
RE
. 
Vorinostat, a histone deacetylase inhibitor, enhances the response of human tumor cells to ionizing radiation through prolongation of gamma-H2AX foci
.
Mol Cancer Ther
2006
;
5
:
1967
74
.
25.
Gaze
MN
,
Chang
YC
,
Flux
GD
,
Mairs
RJ
,
Saran
FH
,
Meller
ST
. 
Feasibility of dosimetry-based high-dose 131I-meta-iodobenzylguanidine with topotecan as a radiosensitizer in children with metastatic neuroblastoma
.
Cancer Biother Radiopharm
2005
;
20
:
195
9
.
26.
Corvi
R
,
Savelyeva
L
,
Schwab
M
. 
Patterns of oncogene activation in human neuroblastoma cells
.
J Neurooncol
1997
;
31
:
25
31
.
27.
Maris
JM
,
Matthay
KK
. 
Molecular biology of neuroblastoma
.
J Clin Oncol
1999
;
17
:
2264
79
.
28.
Bell
E
,
Chen
L
,
Liu
T
,
Marshall
GM
,
Lunec
J
,
Tweddle
DA
. 
MYCN oncoprotein targets and their therapeutic potential
.
Cancer Lett
2010
;
293
:
144
57
.
29.
Quach
A
,
Ji
L
,
Mishra
V
,
Sznewajs
A
,
Veatch
J
,
Huberty
J
, et al
Thyroid and hepatic function after high-dose 131I-metaiodobenzylguanidine (131I-MIBG) therapy for neuroblastoma
.
Pediatr Blood Cancer
2011
;
56
:
191
201
.