Dihydropyrimidine dehydrogenase (DPD) is the rate-limiting enzyme in the catabolism of 5-fluorouracil (5-FU) and its activity is closely associated with cellular sensitivity to 5-FU. This study examines the role of DPD in the antiproliferative effects of 5-FU combined with IFN-α on hepatocellular carcinoma (HCC) cells in culture and asks whether IFN-α could affect DPD expression. The combined action of IFN-α and 5-FU on three HCC lines was quantified by a combination index method. Coadministration of IFN-α and 5-FU showed synergistic effects against HAK-1A and KYN-2 but antagonistic effects against KYN-3. The cellular expression levels of DPD mRNA and protein were markedly up-regulated in KYN-3 cells by IFN-α but were down-regulated in HAK-1A and KYN-2. The expression of thymidylate synthase mRNA and protein was down-regulated by IFN-α in all three cell lines. Coadministration of a selective DPD inhibitor, 5-chloro-2,4-dihydroxypyridine (CDHP), enhanced the antiproliferative effect of 5-FU and IFN-α on KYN-3 ∼4-fold. However, the synergistic effects of 5-FU and IFN-α on HAK-1A and KYN-2 were not affected by CDHP. The antiproliferative effect of 5-FU could thus be modulated by IFN-α, possibly through DPD expression, in HCC cells. Inhibition of DPD activity by CDHP may enhance the efficacy of IFN-α and 5-FU combination therapy in patients with HCC showing resistance to this therapy. [Mol Cancer Ther 2007;6(8):2310–8]

5-Fluorouracil (5-FU) is widely used in the treatment of various gastrointestinal cancers and other types of tumor. It is converted to the active metabolite 5-fluoro-2′-deoxyuridine-5′-monophosphate (FdUMP) and inhibits thymidylate synthase (TS) activity competitively through the formation of a ternary complex of FdUMP, TS, and 5,10-methylenetetrahydrofolate. Cancer cells with high levels of FdUMP and low levels of TS are thus known to be sensitive to 5-FU (1).

Dihydropyrimidine dehydrogenase (DPD) is a rate-limiting enzyme involved in the degradation of pyrimidine bases and pyrimidine-based antimetabolites, such as 5-FU, and so diminishes the antitumor activity of 5-FU. This catabolism occurs mainly in the liver. DPD activity shows wide variation in both cancer patients and the healthy population (2).

The human DPD gene (DPYD) is located on chromosome 1p22. It is a single copy 950-kb gene comprising 23 exons (3), in which 39 mutations and polymorphisms have been identified (46). Abnormalities of DPYD that decrease DPD activity are observed in 3% to 5% of the total population (7), and several patients with congenital DPD deficiency were reported as suffering from severe toxicity after the administration of 5-FU (8). DPD activity in tumor cells is critical to the antitumor effects of 5-FU (9), and its inhibition is expected to enhance these effects.

Hepatocellular carcinoma (HCC) is the fifth most common malignancy in the world. The most effective treatment for patients with HCC is the surgical resection of hepatic lesions. Local therapeutic approaches, such as transcatheter arterial embolization (10), percutaneous transhepatic ethanol injection (11), microwave coagulation (12), and radiofrequency ablation (13), are also effective. However, these therapies are not sufficient for patients with advanced HCC, for whom surgery is often not suitable and whose 5-year survival rate is extremely low (14). For patients with advanced HCC, the clinical response of almost every anticancer drug is insufficient, and several combination chemotherapies have been tried. Combined chemotherapy with 5-FU and IFN-α has been used previously for patients with advanced HCC, with improved therapeutic effects (1517). However, these reports of positive effects are contradicted by a previously observed lack of antitumor activity accompanied by increased toxicity (18). To improve the therapeutic index of 5-FU and IFN-α combination therapy, it is therefore important to establish the cause of this occasional decrease in efficacy and increase in side effects.

Plausible mechanisms, such as an increase in FdUMP, TS inhibition rate, and thymidine phosphorylase activity, a decrease in TS levels, and altered 5-FU pharmacokinetics, have been suggested to explain the improved therapeutic effects of IFN-α and 5-FU (1923). Takaoka et al. (24) showed that transcription of the p53 gene is induced by IFN-α/IFN-β, accompanied by an increase in p53 protein level, and that the apoptotic response by IFN-β combined with 5-FU was enhanced. Milano et al. (25) reported that IFN-α inhibits DPD activity in human tumor cells, suggesting that inhibition of DPD activity could be involved in 5-FU–induced antiproliferative activity. Consensus IFN was shown to enhance the antiproliferative effect of 5-FU against hepatoma cells through down-regulation of DPD expression (23). By contrast, increased expression of DPD protein by IFN-γ was reportedly observed at a concentration equivalent to that in the sera of patients (26).

According to the presence or absence of synergism by the combination of 5-FU and IFN-α, we classified six human HCC cell lines into two groups: the S-group containing three cell lines, which showed a synergistic effect, and the A-group containing the remaining three cell lines, which showed additive effects (27). The expression levels of type I IFN receptor subunits were specifically up-regulated by 5-FU in all three cell lines of the S-group but not in those of the A-group (27). In this study, we asked whether DPD could limit the antiproliferative effect of 5-FU against HCC cells in culture when 5-FU was applied in combination with IFN-α. This work also shows that inhibiting DPD activity in HCC cells with high DPD levels improves the efficacy of 5-FU combined with IFN-α, following up-regulation by IFN-α.

Drugs

5-FU was purchased from Kyowa Hakko Kogyo Co. Ltd. (5-FU Injection 250 Kyowa) and natural human IFN-α was purchased from Otsuka Pharmaceutical Co. Ltd. (OIF). 5-Chloro-2,4-dihydroxypyridine (CDHP) was a gift from Taiho Pharmaceutical Co. Ltd.

Cell Lines

HCC cell lines, KYN-2, KYN-3, and HAK-1A (28, 29), were grown in DMEM (Nissui Seiyaku Co.) with 10% fetal bovine serum (FetalClone III, Hyclone) in a humidified atmosphere of 5% CO2 at 37°C. We confirmed the expression of type I IFN receptor subunits 1 and 2 in these three HCC cell lines (27).

Cytotoxicity Tests

Cells were seeded into 96-well plates at 1,000 cells/100 μL/well and incubated overnight. On the following day, 100-μL aliquots containing IFN-α and/or 5-FU with or without CDHP were added to each well and cells were cultured for a further 5 days. The number of viable cells was estimated by assaying the activity of cellular succinate dehydrogenases using WST-8 reagent (Cell Counting Kit-8, Dojindo; ref. 30). We confirmed that untreated groups of KYN-2, KYN-3, and HAK-1A cells grew exponentially for 6 days under these experimental conditions (data not shown).

Combination Index Analysis

The combined effects of 5-FU and IFN-α were quantified using a combination index (CI) method developed by Chou and Talalay (31). This method involves plotting dose-effect curves, for each agent and their combination, using the median-effect equation: fa / fu = (D / Dm)m, where D is the dose of the drug, Dm is the dose required for a 50% effect (equivalent to IC50), fa and fu are the affected and unaffected fractions, respectively (fa = 1 − fu), and m is the exponent signifying the sigmoidicity of the dose-effect curve.

In this study, relative concentrations (RC) of IFN-α and 5-FU, determined as (concentration) / (IC50 value), were used for analysis. The computer software Xlfit version 2.0.6 (ID Business Solutions Ltd.) was used to calculate the values of Dm and m. The CI used for the analysis of the drug combinations was determined by the isobologram equation for mutually nonexclusive drugs that have different modes of action: CI = (D)1 / (Dx)1 + (D)2 / (Dx)2 + (D)1(D)2 / (Dx)1(Dx)2, where (D)1 and (D)2 are RCs of drugs 1 and 2 and x is the percentage of inhibition. Combination indices CI < 1, CI = 1, and CI > 1 indicate synergism, additive effects, and antagonism, respectively.

Quantitative Real-time Reverse Transcription-PCR

Total RNA was extracted using Isogen (Nippon Gene Co., Ltd.) and reverse transcribed using a reverse-transcription system (Promega Corp.) according to the manufacturer's instructions. Quantitative real-time reverse transcription-PCR was done with an ABI Prism 7300 (PE Applied Biosystems). The primers used were as follows: TS 5′-GAATCACATCGAGCCACTGAAA-3′ (forward primer), 5′-CAGCCCAACCCCTAAAGACTGA-3′ (reverse primer), and 5′-(FAM)TTCAGCTTCAGCGAGAACCCAGA(TAMRA)-3′ (probe) and DPD 5′-AATGATTCGAAGAGCTTTTGAAGC-3′ (forward primer), 5′-GTTCCCCGGATGATTCTGG-3′ (reverse primer), and 5′-(FAM)TGCCCTCACCAAAACTTTCTCTCTTGATAAGGA(TAMRA)-3′ (probe). Primers and Taqman probes for glyceraldehyde-3-phosphate dehydrogenase were prepared by Assay-on-Demand Gene Expression Products (PE Applied Biosystems).

Western Blotting

HCC cells were cultured for 48 h with 0, 20, 100, or 500 IU/mL IFN-α. Total protein was extracted using a protein extraction reagent (M-PER, Pierce) supplemented with protease inhibitors (Halt Protease Inhibitor Cocktail kit, Pierce). Cell lysates were loaded into 7.5% SDS-polyacrylamide gels. After electrophoresis, the separated proteins were electrotransblotted onto polyvinylidene difluoride membranes (Immobilon-P membrane, Millipore). After blocking, membranes were probed with antihuman DPD polyclonal rabbit antibody and antihuman TS monoclonal mouse antibody (gifts from Taiho Pharmaceutical). The proteins were visualized using horseradish peroxidase–conjugated antibodies (Pierce) followed by enhanced chemiluminescence (Pierce). The intensity of luminescence was quantified using an image analysis system (LAS-1000, Fuji Film).

Enzyme Assay for DPD Activity

DPD activity was measured using [6-14C]5-FU as a substrate (32, 33). Cells were homogenized and centrifuged at 105,000 × g for 60 min at 4°C, and the supernatant was used for assay. A reaction mixture containing 10 mmol/L potassium phosphate (pH 8.0), 0.5 mmol/L EDTA, 0.5 mmol/L 2-mercaptoethanol, 2 mmol/L DTT, 5 mmol/L MgCl2, 20 μmol/L [6-14C]5-FU, 0.1 mmol/L NADPH, and 25 μL of cell extract in a total volume of 50 μL was incubated at 37°C for 30 min. After chemical hydrolyzation and neutralization using KOH and HClO4, a 5-μL aliquot was applied to a TLC plate (2.5 × 20 cm, silica gel 60 F254 plate, Merck) and developed with a mixture of ethanol and 1 mol/L ammonium acetate (5:1, v/v) and diethylether, acetone, chloroform, and water (50:50:40:1, v/v). DPD activity was determined as the sum of the products converted from 5-FU (i.e., dihydrouracil, 2-fluoro-β-ureidopropionic acid, and 2-fluoro-β-alanine) that were visualized and quantified with an imaging analyzer (BAS-2000, Fujix).

Comparison of Drug Sensitivity and mRNA Levels of DPD and TS in Three HCC Cell Lines

The sensitivities of three HCC lines, HAK-1A, KYN-2, and KYN-3, to separately administered 5-FU and IFN-α were determined as IC50 values. KYN-3 was the most resistant to 5-FU of the three HCC lines. The IC50 values of IFN-α in HAK-1A, KYN-2, and KYN-3 cells were 720, 510, and 24 IU/mL, respectively. KYN-3 cellular sensitivity to IFN-α was therefore approximately 30-fold and 20-fold higher than in HAK-1A and KYN-2 cell lines, respectively. The IC50 value of IFN-α in HepG2 cells established from human hepatoblastoma and widely used in experiments was found to be over 10,000 IU/mL (data not shown).

Sensitivity was then compared with mRNA levels of DPD and TS, and DPD activity (as conversion rate from [6-14C]5-FU to its metabolites) in the three cell lines, which were shown to be broadly comparable (Table 1). DPD mRNA levels relative to those of KYN-3 cells, taken as 100%, and those of TS relative to HAK-1A, taken as 100%, are shown in Table 1. The cellular level of TS mRNA in KYN-2 cells was much lower than in the other two lines. Basal DPD activity in KYN-3 cells was approximately 5.5-fold and 9.1-fold higher than in HAK-1A and KYN-2 cell lines, respectively.

Table 1.

HCC cell sensitivities to 5-FU and IFN-α, DPD, and TS mRNA expression levels and DPD activities

Cell lineIC50*
Relative mRNA levels
DPD activity (pmol/min/mg protein)
5-FU (μmol/L)IFN-α (IU/mL)DPDTS
HAK-1A 2.1 720 15 100 2.8 ± 0.9 
KYN-2 1.8 510 23 14 1.7 ± 0.7 
KYN-3 9.8 24 100 46 15.5 ± 1.6 
Cell lineIC50*
Relative mRNA levels
DPD activity (pmol/min/mg protein)
5-FU (μmol/L)IFN-α (IU/mL)DPDTS
HAK-1A 2.1 720 15 100 2.8 ± 0.9 
KYN-2 1.8 510 23 14 1.7 ± 0.7 
KYN-3 9.8 24 100 46 15.5 ± 1.6 
*

The IC50 value that caused 50% growth inhibition was calculated from the log-logit regression line. The assays were carried out in quadruplicate. Experiments were repeated twice with essentially similar results.

DPD mRNA levels are shown relative to those of KYN-3 cells, taken as 100%, and TS mRNA levels are shown relative to those of HAK-1A cells, taken as 100%.

Determinations were carried out in triplicate and data represent mean ± SD.

Quantitative Analysis of the Combination Effects of 5-FU and IFN-α

Dose-response curves of 5-FU alone and in combination with various concentrations of IFN-α are shown in Fig. 1A to C. Because sensitivities to IFN-α and 5-FU alone were different for each HCC cell line, RCs to the IC50 value were used. As the concentration of combined IFN-α was increased, the dose-response curves of 5-FU were shifted down in an IFN-α concentration-dependent manner in all three HCC cell lines. For instance, following cotreatment with 312 IU/mL (RC = 0.45) IFN-α,the dose-response curve of HAK-1A to 5-FU was significantly shifted down and the 5-FU IC50 value of 2.3 μmol/L was reduced to 0.28 μmol/L. However, there were clear differences between the three HCC cell lines. For KYN-3 cells, after treatment with the equivalent RC, 0.42 (8.0 IU/mL) IFN-α,the dose-response curve of 5-FU was significantly shifted down, but the 5-FU IC50 value of 9.6 μmol/L was only reduced to 5.5 μmol/L.

Figure 1.

Antiproliferative effects of 5-FU and IFN-α on HAK-1A (A), KYN-2 (B), and KYN-3 (C) cells. Assays were carried out independently in quadruplicate. Points, mean; bars, SD. †, RC = (concentration) / (IC50 value). In this experiment, IC50 value of IFN-α against HAK-1A, KYN-2, and KYN-3 was 700, 490, and 20 IU/mL, respectively. ‡, EF = 1 / [(RC of IFN-α) + (RC of 5-FU)]. EF > 1, synergistic; EF = 1, additive; EF < 1, antagonistic. Experiments were repeated twice with similar results.

Figure 1.

Antiproliferative effects of 5-FU and IFN-α on HAK-1A (A), KYN-2 (B), and KYN-3 (C) cells. Assays were carried out independently in quadruplicate. Points, mean; bars, SD. †, RC = (concentration) / (IC50 value). In this experiment, IC50 value of IFN-α against HAK-1A, KYN-2, and KYN-3 was 700, 490, and 20 IU/mL, respectively. ‡, EF = 1 / [(RC of IFN-α) + (RC of 5-FU)]. EF > 1, synergistic; EF = 1, additive; EF < 1, antagonistic. Experiments were repeated twice with similar results.

Close modal

An enhancement factor (EF) was defined to evaluate synergism between 5-FU and IFN-α, based on a 50% antiproliferative effect, as EF = 1 / [(RC of IFN-α) + (RC of 5-FU)]. When EF is 1, this combined effect is additive; values >1 or <1 imply synergistic or antagonistic effects, respectively. EF values of HAK-1A and KYN-2 were >1 at almost all combined doses, but EF values of KYN-3 at all tested combined doses were closer to 1. The combined effect of 5-FU and IFN-α on HAK-1A and KYN-2 cells was thus judged to be synergistic and that on KYN-3 to be additive, consistent with our previous study (27).

The synergism of IFN-α and 5-FU at various fractional efficacies was then quantified using the CI methods of Chou and Talalay (31). The antiproliferative effects of 5-FU on HAK-1A and KYN-2 cells were synergistically enhanced by IFN-α, indicated by CI values of <1 at most fractional effects (Fig. 2A and B). By contrast, the CI value in KYN-3 cells was >1 for fractional effects of 0.7 and under (Fig. 2C). The combined effect of IFN-α and 5-FU against KYN-3 thus seems to be antagonistic, except at very high fractional effect levels. These data show that the degree of synergism between IFN-α and 5-FU against KYN-3 cells (with a higher CI value) is weaker than against HAK-1A and KYN-2 cells over a large range of fractional effects.

Figure 2.

Quantitative analysis of synergy between 5-FU and IFN-α against HAK-1A (A), KYN-2 (B), and KYN-3 (C) cells using the CI method. CI > 1, antagonism; CI = 1, additive; CI < 1, synergistic. †, ratio of RC = (concentration of 5-FU) / (IC50 of 5-FU):(concentration of IFN-α) / (IC50 of IFN-α).

Figure 2.

Quantitative analysis of synergy between 5-FU and IFN-α against HAK-1A (A), KYN-2 (B), and KYN-3 (C) cells using the CI method. CI > 1, antagonism; CI = 1, additive; CI < 1, synergistic. †, ratio of RC = (concentration of 5-FU) / (IC50 of 5-FU):(concentration of IFN-α) / (IC50 of IFN-α).

Close modal

Altered Expression of mRNA and Protein of TS and DPD by IFN-α

Cellular mRNA levels of DPD and TS were examined following treatment with or without 500 IU/mL IFN-α for 24 h (Fig. 3A and B). IFN-α treatment resulted in an ∼7.5-fold increase in DPD mRNA levels in KYN-3 cells while reducing DPD mRNA levels in HAK-1A and KYN-2 cells to approximately one quarter and one third of that in IFN-α untreated cells, respectively (Fig. 3A). TS mRNA levels in all three HCC cell lines decreased after treatment with IFN-α to ∼60% of those in untreated cells (Fig. 3B).

Figure 3.

Effects of 500 IU/mL IFN-α on DPD (A) and TS (B) mRNA and protein expression levels in HAK-1A, KYN-2, and KYN-3 cells. Expression levels were measured by quantitative real-time reverse transcription-PCR and normalized to glyceraldehyde-3-phosphate dehydrogenase. The fold increases are shown relative to the initial level, taken as 1.0. Determinations were carried out in triplicate. Dotted and black columns, mean value of relative mRNA levels in HCC cells untreated and treated with IFN-α, respectively; bars, SD. *, P < 0.05; **, P < 0.01; ***, P < 0.001, differences are statistically significant by Welch's test, compared with untreated groups.

Figure 3.

Effects of 500 IU/mL IFN-α on DPD (A) and TS (B) mRNA and protein expression levels in HAK-1A, KYN-2, and KYN-3 cells. Expression levels were measured by quantitative real-time reverse transcription-PCR and normalized to glyceraldehyde-3-phosphate dehydrogenase. The fold increases are shown relative to the initial level, taken as 1.0. Determinations were carried out in triplicate. Dotted and black columns, mean value of relative mRNA levels in HCC cells untreated and treated with IFN-α, respectively; bars, SD. *, P < 0.05; **, P < 0.01; ***, P < 0.001, differences are statistically significant by Welch's test, compared with untreated groups.

Close modal

Cellular protein levels of DPD and TS were also determined by Western blot analysis. The intensities of blotted bands were normalized by that of β-actin, and fold increase was measured as the relative intensity to that of untreated cells, taken as 1.0. Protein extracts of 10 μg/lane were loaded for KYN-3 cells, and 50 μg/lane were loaded for both HAK-1A and KYN-2 cells (Fig. 4). DPD protein levels were roughly comparable with mRNA levels and DPD activity in all three cell lines. Expression of DPD protein, molecular weight of 110,000, was down-regulated in HAK-1A and KYN-2 cells when treated with over 100 IU/mL IFN-α for 48 h. Conversely, that in KYN-3 cells was up-regulated after treatment with over 20 IU/mL IFN-α for 48 h in a concentration-dependent manner (Fig. 4). Expression of TS protein, molecular weight of 36,000, was down-regulated to a similar extent in a concentration-dependent manner in all three HCC cell lines when treated with IFN-α.

Figure 4.

Protein expression levels of DPD and TS in HAK-1A, KYN-2, and KYN-3 cells treated with IFN-α. Protein expressions in HCC cells treated with IFN-α for 48 h were measured by Western blotting. Protein extracts of 10 μg/lane were loaded for KYN-3 cells, and 50 μg/lane were loaded for both HAK-1A and KYN-2 cells. Experiments were repeated twice with similar results. The intensities of immunoblotted bands were quantified by image analyzing methods. The fold increases relative to the initial level, taken as 1.0, are shown under the bands.

Figure 4.

Protein expression levels of DPD and TS in HAK-1A, KYN-2, and KYN-3 cells treated with IFN-α. Protein expressions in HCC cells treated with IFN-α for 48 h were measured by Western blotting. Protein extracts of 10 μg/lane were loaded for KYN-3 cells, and 50 μg/lane were loaded for both HAK-1A and KYN-2 cells. Experiments were repeated twice with similar results. The intensities of immunoblotted bands were quantified by image analyzing methods. The fold increases relative to the initial level, taken as 1.0, are shown under the bands.

Close modal

Effect of a DPD Selective Inhibitor on the Antiproliferative Effect of 5-FU and IFN-α

The combined antiproliferative effects of 5-FU and IFN-α can be modulated by treatment with a DPD competitive inhibitor, CDHP. We first examined the effect of CDHP on DPD activity in three HCC cell lines. KYN-3 cell extracts showed relatively high DPD activity of ∼15 pmol/min/mg protein (consistent with Table 1 findings). By contrast, DPD activity in HAK-1A and KYN-2 cells was very low with a marginal limit of evaluation for enzyme activity. DPD activity in KYN-3 cells was reduced to 40%, 7%, and 5% of basal activity after treatment with 0.7, 7.0, and 70 μmol/L CDHP, respectively (Fig. 5). There seemed to be almost no marked inhibition of DPD activity by CDHP in the other two cell lines, possibly due to their low enzyme activities.

Figure 5.

Inhibitory effect of CDHP on DPD activity. DPD activity was measured using [6-14C]5-FU as a substrate. Residual [6-14C]5-FU and metabolites were separated by TLC and visualized with an imaging analyzer (A). Each sample was developed on separated and independent TLC plate. Assays were carried out in triplicate. Columns, DPD; bars, SD (B). *, P < 0.001, differences are statistically significant by Welch's test, compared with CDHP untreated group.

Figure 5.

Inhibitory effect of CDHP on DPD activity. DPD activity was measured using [6-14C]5-FU as a substrate. Residual [6-14C]5-FU and metabolites were separated by TLC and visualized with an imaging analyzer (A). Each sample was developed on separated and independent TLC plate. Assays were carried out in triplicate. Columns, DPD; bars, SD (B). *, P < 0.001, differences are statistically significant by Welch's test, compared with CDHP untreated group.

Close modal

We next examined whether cotreatment of CDHP could modulate the antiproliferative effect of 5-FU alone and in combination with IFN-α in three HCC cell lines. As seen in Fig. 6A to C, the dose-response curves of 5-FU alone in HAK-1A, KYN-2, and KYN-3 cells and those of 5-FU combined with IFN-α in HAK-1A and KYN-2 cells were not shifted by cotreatment with CDHP. None of these differences were significant at all data points. By contrast, there seemed a marked and significant shift of the 5-FU and IFN-α combined dose-response curve in KYN-3 cells to CDHP: the 5-FU IC50 value of 4.6 μmol/L was reduced to 1.1 μmol/L by cotreatment with CDHP in the presence of IFN-α (Fig. 6C).

Figure 6.

Effect of CDHP on combined antiproliferative effect of IFN-α and 5-FU in HAK-1A (A), KYN-2 (B), and KYN-3 (C) cells. Assays were carried out in quadruplicate. Experiments were repeated twice with similar results. IC50 values of 5-FU against HCC cells treated with 5-FU alone (†), 5-FU/CDHP (‡), 5-FU/IFN-α (§), and 5-FU/IFN-α/CDHP (¶).

Figure 6.

Effect of CDHP on combined antiproliferative effect of IFN-α and 5-FU in HAK-1A (A), KYN-2 (B), and KYN-3 (C) cells. Assays were carried out in quadruplicate. Experiments were repeated twice with similar results. IC50 values of 5-FU against HCC cells treated with 5-FU alone (†), 5-FU/CDHP (‡), 5-FU/IFN-α (§), and 5-FU/IFN-α/CDHP (¶).

Close modal

Consistent with the findings of our recent study (27), the combination of 5-FU and IFN-α showed a synergistic antiproliferation effect on two HCC cell lines, HAK-1A and KYN-2, and an additive or antagonistic antiproliferation effect on KYN-3 cells (Figs. 1 and 2). Expression of type 1 IFN receptor was specifically up-regulated by exposure to 5-FU in both HAK-1A and KYN-2, but not KYN-3 cells, suggesting that the modulation of IFN receptor expression by 5-FU could play a pivotal role in therapeutic efficacy (27). In this study, we further examined the molecular events underlying the antiproliferative effects of 5-FU and IFN-α. One of the major mechanisms of antiproliferative activity of 5-FU is the inhibition of TS activity with formation of the ternary complex of FdUMP, TS, and 5,10-methylenetetrahydrofolate. However, we observed a marked IFN-α−induced decrease in TS expression at similar levels in all three cell lines, suggesting that modulation of TS expression itself might not be directly involved in the absence or presence of synergism by the combination of 5-FU and IFN-α.

DPD is a key enzyme involved in 5-FU inactivation, which modulates FdUMP levels and controls formation of the ternary complex. Several clinical studies have shown that the intratumoral expression level of DPD is closely associated with clinical responses to 5-FU in patients with colorectal cancer (1), gastric cancer (34), and non–small cell lung cancer (35). In our present study, expression of DPD protein and mRNA levels in KYN-3 cells were specifically increased 5-fold or more over the basal level after exposure to IFN-α (Figs. 3 and 4). By contrast, down-regulation of DPD by IFN-α was observed in both HAK-1A and KYN-2.

In these two HCC cell lines, we previously proposed that 5-FU–induced up-regulation of the IFN receptor was the main mechanism underlying the synergistic antiproliferative effect of 5-FU and IFN-α (27). Moreover, down-regulation of DPD by IFN-α in these two cell lines might be involved in the synergistic effect. By contrast, up-regulation of DPD by IFN-α might account for the antagonism between IFN-α and 5-FU in KYN-3 cells.

Shestopal et al. (36) reported that 5′ flanking region of DPYD gene lacks the canonical TATA and CCAAT boxes, however, contains several cis-acting regulatory elements including binding sites for activator protein-2, nuclear factor-κB, Sp1, and Egr families. About the regulatory mechanism for IFN-α modulation of DPYD expression, we analyzed the sequence of 1.2 kb of 5′ flanking region of DPYD and found that this region contains two putative consensus binding sites for signal transducer and activator of transcription families (data not shown), suggesting that gene expression of DPYD is highly susceptible to IFN-α, which strongly activates signal transducer and activator of transcription 1 and signal transducer and activator of transcription 2 through the Janus-activated kinase-signal transducer and activator of transcription pathway. However, it remains unclear why DPYD expression is differentially controlled by IFN-α in HCC cell lines. Further elucidation of this differential regulatory mechanism at the molecular basis is required.

Our present study showed that cotreatment with the DPD inhibitor CDHP further synergistically enhanced the antiproliferative effect of 5-FU and IFN-α in KYN-3 cells only and not in HAK-1A and KYN-2 cells (Fig. 6). The antiproliferative effect of 5-FU alone was only slightly altered by CDHP cotreatment in KYN-3 cells, if any, and not at all in the other two cell lines (Fig. 6). This IC50 reduction, however, was not statistically significant. Basal DPD activity and expression levels were much higher in KYN-3 cells than in HAK-1A and KYN-2 cells. Moreover, cellular DPD levels were specifically up-regulated >5-fold in KYN-3 cells by IFN-α, but this was not observed in HAK-1A and KYN-2 cells. It was presumed that 15 pmol/min/mg protein of basal DPD activity in KYN-3 cells was up-regulated to 80 to 110 pmol/min/mg protein. Taken together, this suggests that a relatively high DPD activity might be more susceptible to inhibition by CDHP, resulting in an apparent synergistic effect of CDHP on the antiproliferative effect by 5-FU and IFN-α.

Certain levels of DPD in cancer cells could be sensitive to CDHP-induced inhibition. A relevant study by Takechi et al. (37) showed that the antiproliferative activity of 5-FU could be markedly enhanced by cotreatment with 69 μmol/L CDHP in two human tumor cell lines with relatively high DPD activities, approximately 101 and 153 pmol/min/mg protein, respectively, but not those with low enzyme activity, 33 pmol/min/mg protein. However, this plausible mechanism why CDHP did induce synergism of 5-FU and IFN-α against only KYN-3 cells requires further study to validate these findings.

CDHP has been applied as a modulator in the newly developed antimetabolite TS-1 (Taiho Pharmaceutical). TS-1 consists of tegafur, CDHP, and potassium oxonate in a molar ratio of 1:0.4:1 (38). Potassium oxonate is a competitive inhibitor for orotate phosphoribosyltransferase that activates 5-FU. Potassium oxonate is mainly distributed in the gastrointestinal tract after p.o. administration and prevents gastrointestinal toxicity induced by 5-FU without reducing 5-FU activity in tumor (38, 39). In Japan, TS-1 has been used to treat patients with gastric, head and neck, and pancreatic cancers and shows potent therapeutic efficacy against gastric tumors, with a response rate of 46.5% (4042). Nakamura et al. (43) applied a new combination regimen of TS-1 and IFN-α to advanced HCC patients with portal vein thrombus and multiple pulmonary metastases and observed some improvement in therapeutic efficacy. The combination of TS-1 and IFN-α could therefore be effective against patients with advanced HCC. However, the side effects of these combination therapies should be seriously considered before their implementation. Further studies are required to determine how DPD could be differentially controlled between normal cells including normal hepatic cells and malignant hepatic cells in patients with HCC when DPD inhibitory drugs are introduced.

Grant support: Health and Labour Sciences Research grants of Third Term Comprehensive Control Research for Cancer from the Ministry of Health, Labour and Welfare, Japan and the 21st Century COE Program for Medical Sciences, Kurume University, supported by the Ministry of Education, Culture, Sports, Science and Technology, Japan.

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.

We thank T. Kobunai, H. Tsujimoto, J. Chikamoto, Y. Fukui, Dr. Y. Basaki, and Dr. M. Kiniwa for technical support and fruitful discussion and N. Shinbaru for editorial help.

1
Salonga D, Danenberg KD, Johnson M, et al. Colorectal tumors responding to 5-fluorouracil have low gene expression levels of dihydropyrimidine dehydrogenase, thymidylate synthase, and thymidine phosphorylase.
Clin Cancer Res
2000
;
6
:
1322
–7.
2
Etienne MC, Lagrange JL, Dassonville O, et al. Population study of dihydropyrimidine dehydrogenase in cancer patients.
J Clin Oncol
1994
;
12
:
2248
–53.
3
Wei X, Elizondo G, Sapone A, et al. Characterization of the human dihydropyrimidine dehydrogenase gene.
Genomics
1998
;
51
:
391
–400.
4
van Kuilenburg ABP. Dihydropyrimidine dehydrogenase and efficacy and toxicity of 5-fluorouracil.
Eur J Cancer
2004
;
40
:
939
–50.
5
Raida M, Schwabe W, Hausler P, et al. Prevalence of common point mutation in the dihydropyrimidine dehydrogenase (DPD) gene within the 5′-splice donor site of intron 14 in patients with severe 5-fluorouracil (5-FU)-related toxicity compared with controls.
Clin Cancer Res
2001
;
7
:
2832
–9.
6
van Kuilenburg ABP, Meinsma JR, Zoetekouw L, van Gennip AH. High prevalence of the IVS14+1G>A mutation in the dihydropyrimidine dehydrogenase gene of patients with severe 5-fluorouracil-associated toxicity.
Pharmacogenetics
2002
;
12
:
555
–8.
7
Johnson MR, Diasio RB. Importance of dihydropyrimidine dehydrogenase (DPD) deficiency in patients exhibiting toxicity following treatment with 5-fluorouracil.
Adv Enzyme Regul
2001
;
41
:
151
–7.
8
van Kuilenburg ABP, Muller EW, Haasjes J, et al. Lethal outcome of a patient with a complete dihydropyrimidine dehydrogenase (DPD) deficiency after administration of 5-fluorouracil: frequency of the common IVS14+1G>A mutation causing DPD deficiency.
Clin Cancer Res
2001
;
7
:
1149
–53.
9
Allegra CJ. Dihydropyrimidine dehydrogenase activity: prognostic partner of 5-fluorouracil?
Clin Cancer Res
1999
;
5
:
1947
–9.
10
Cheung YC, Ko SF, Ng SH, Chan SC, Cheng YF. Survival outcome of lobar or segmental transcatheter arterial embolization with ethanol-lipiodol mixture in treating hepatocellular carcinoma.
World J Gastroenterol
2005
;
11
:
2792
–5.
11
Ebara M, Okabe S, Kita K, et al. Percutaneous ethanol injection for small hepatocellular carcinoma: therapeutic efficacy based on 20-year observation.
J Hepatol
2005
;
43
:
458
–64.
12
Kawamoto C, Ido K, Isoda N, et al. Long-term outcomes for patients with solitary hepatocellular carcinoma treated by laparoscopic microwave coagulation.
Cancer
2005
;
103
:
985
–93.
13
Tateishi R, Shiina S, Teratani T, et al. Percutaneous radiofrequency ablation for hepatocellular carcinoma. An analysis of 1000 cases.
Cancer
2005
;
103
:
1201
–9.
14
Llovet JM. Updated treatment approach to hepatocellular carcinoma.
J Gastroenterol
2005
;
40
:
225
–35.
15
Patt YZ, Hassan MM, Lozano RD, et al. Phase II trial of systemic continuous fluorouracil and subcutaneous recombinant interferon Alfa-2b for treatment of hepatocellular carcinoma.
J Clin Oncol
2003
;
21
:
421
–7.
16
Hosogi H, Ikai I, Hatano E, et al. Complete response by a combination of 5-fluorouracil and interferon-α chemotherapy for lung metastasis of hepatocellular carcinoma after hepatic resection with portal and hepatic vein tumor thrombectomy.
Hepatol Res
2005
;
33
:
320
–4.
17
Ohmoto K, Iguchi Y, Mimura N, et al. Combined intraarterial 5-fluorouracil and intramuscular interferon-α therapy for advanced hepatocellular carcinoma.
Hepatogastroenterology
2003
;
50
:
1780
–2.
18
Stuart K, Tessitore J, Huberman M. 5-Fluorouracil and α-interferon in hepatocellular carcinoma.
Am J Clin Oncol
1996
;
19
:
136
–9.
19
Ismail A, Van Groeningen CJ, Hardcastle A, et al. Modulation of fluorouracil cytotoxicity by interferon-α and -γ.
Mol Pharmacol
1998
;
53
:
252
–61.
20
Schwartz EL, Hoffman M, O'Connor CJ, Wadler S. Stimulation of 5-fluorouracil metabolic activation by interferon-α in human colon carcinoma cells.
Biochem Biophys Res Commun
1992
;
182
:
1232
–9.
21
Yee LK, Allegra CJ, Steinberg SM, Grem JL. Decreased catabolism of 5-fluorouracil in peripheral blood mononuclear cells during therapy with 5-fluorouracil, leucovorin and interferon α-2a.
J Natl Cancer Inst
1992
;
84
:
1820
–5.
22
Yao Y, Kubota T, Sato K, Takeuchi H, Kitai R, Matsukawa S. Interferons upregulate thymidine phosphorylase expression via JAK-STAT-dependent transcriptional activation and mRNA stabilization in human glioblastoma cells.
J Neurooncol
2005
;
72
:
217
–23.
23
Dou J, Iwashita Y, Sasaki A, et al. Consensus interferon enhances the anti-proliferative effect of 5-fluorouracil on human hepatoma cells via downregulation of dihydropyrimidine dehydrogenase expression.
Liver Int
2005
;
25
:
148
–52.
24
Takaoka A, Hayakawa S, Yanai H, et al. Integration of interferon-α/β signaling to p53 responses in tumour suppression and antiviral defence.
Nature
2003
;
424
:
516
–23.
25
Milano G, Fischel JL, Etienne MC, et al. Inhibition of dihydropyrimidine dehydrogenase by α-interferon: experimental data on human tumor cell lines.
Cancer Chemother Pharmacol
1994
;
34
:
147
–52.
26
Miwa M, Kojima T, Naruse T. Serum factors attenuating the anti-tumor activity of 5-fluorouracil.
Cancer Biother Radiopharm
2001
;
16
:
317
–22.
27
Yano H, Maruiwa M, Murakami T, et al. A new human pleomorphic hepatocellular carcinoma cell line, KYN-2.
Acta Pathol Jpn
1988
;
38
:
953
–66.
28
Yano H, Iemura A, Fukuda K, Mizoguchi A, Haramaki M, Kojiro M. Establishment of two distinct human hepatocellular carcinoma cell lines from a single nodule showing clonal dedifferentiation of cancer cells.
Hepatology
1993
;
18
:
320
–7.
29
Oie S, Ono M, Yano H, et al. The upregulation of type I interferon receptor gene plays a key role in hepatocellular carcinoma cells in the synergistic antiproliferative effect by 5-fluorouracil and interferon-α.
Int J Oncol
2006
;
29
:
1469
–78.
30
Ishiyama M, Miyazono Y, Sasamoto K, Ohkura Y, Ueno K. A highly water-soluble disulfonated tetrazolium salt as a chromogenic indicator for NADH as well as cell viability.
Talanta
1997
;
44
:
1299
–305.
31
Chou TC, Talalay P. Quantitative analysis of dose-effect relationships: the combined effects of multiple drugs or enzyme inhibitors.
Adv Enzyme Regul
1984
;
22
:
2
–55.
32
Fukushima M, Morita M, Ikeda K, Nagayama S. Population study of expression of thymidylate synthase and dihydropyrimidine dehydrogenase in patients with solid tumors.
Int J Mol Med
2003
;
12
:
839
–44.
33
Takechi T, Uchida J, Fujioka A, Fukushima M. Enhancing 5-fluorouracil cytotoxicity by inhibiting dihydropyrimidine dehydrogenase activity with uracil in human tumor cells.
Int J Oncol
1997
;
11
:
1041
–4.
34
Ishikawa Y, Kubota T, Otani Y, et al. Dihydropyrimidine dehydrogenase and messenger RNA levels in gastric cancer: possible predictor for sensitivity to 5-fluorouracil.
Jpn J Cancer Res
2000
;
91
:
105
–12.
35
Huang CL, Yokomise H, Kobayashi S, Fukushima M, Hitomi S, Wada H. Intratumoral expression of thymidylate synthase and dihydropyrimidine dehydrogenase in non-small cell lung cancer patients treated with 5-FU-based chemotherapy.
Int J Oncol
2000
;
17
:
47
–54.
36
Shestopal SA, Johnson MR, Diasio RB. Molecular cloning and characterization of the human dihydropyrimidine dehydrogenase promoter.
Biochim Biophys Acta
2000
;
1494
:
162
–9.
37
Takechi T, Fujioka A, Matsushima E, Fukushima M. Enhancement of the antitumour activity of 5-fluorouracil (5-FU) by inhibiting dihydropyrimidine dehydrogenase activity (DPD) using 5-chloro-2,4-dihydroxypyridine (CDHP) in human tumor cells.
Eur J Cancer
2002
;
38
:
1271
–7.
38
Shirasaka T, Shimamoto Y, Ohshimo H, et al. Development of a novel form of an oral 5-fluorouracil derivative (S-1) directed to the potentiation of the tumor selective cytotoxicity of 5-fluorouracil by two biochemical modulators.
Anticancer Drugs
1996
;
7
:
548
–57.
39
Shirasaka T, Shimamoto Y, Fukushima M. Inhibition by oxonic acid of gastrointestinal toxicity of 5-fluorouracil without loss of its antitumor activity in rats.
Cancer Res
1993
;
53
:
4004
–9.
40
Sugimachi K, Maehara Y, Horikoshi N, et al.; The S-1 Gastrointestinal Cancer Study Group. An early phase II study of oral S-1, a newly developed 5-fluorouracil derivative for advanced and recurrent gastrointestinal cancers.
Oncology
1999
;
57
:
202
–10.
41
Sakata Y, Ohtsu A, Horikoshi N, Sugimachi K, Mitachi Y, Taguchi T. Late phase II study of novel oral fluoropyrimidine anticancer drug S-1 (1 M tegafur-0.4 M gimestat-1 M otastat potassium) in advanced gastric cancer patients.
Eur J Cancer
1998
;
34
:
1715
–20.
42
Koizumi W, Kurihara M, Nakano S, Hasegawa K; For the S-1 Cooperative Gastric Cancer Study Group. Phase II study of S-1, a novel oral derivative of 5-fluorouracil, in advanced gastric cancer.
Oncology
2000
;
58
:
191
–7.
43
Nakamura M, Nagano H, Wada H, et al. A case of hepatocellular carcinoma with multiple lung, spleen, and remnant liver metastasis successfully treated by combination chemotherapy with the novel oral DPD-inhibiting chemotherapeutic drug S-1 and interferon-α.
Gastroenterology
2006
;
41
:
1120
–5.