Abstract
Neuroblastoma is the most common extracranial solid tumor of childhood. The activity of J1 (l-melphalanyl-p-l-fluorophenylalanine ethyl ester), an enzymatically activated melphalan prodrug, was evaluated in neuroblastoma models in vitro and in vivo. Seven neuroblastoma cell lines with various levels of drug resistance were screened for cytotoxicity of J1 alone or in combination with standard cytotoxic drugs, using a fluorometric cytotoxicity assay. J1 displayed high cytotoxic activity in vitro against all neuroblastoma cell lines, with IC50 values in the submicromolar range, significantly more potent than melphalan. The cytotoxicity of J1, but not melphalan, could be significantly inhibited by the aminopeptidase inhibitor bestatin. J1 induced caspase-3 cleavage and apoptotic morphology, had additive effects in combination with doxorubicin, cyclophosphamide, carboplatin, and vincristine, and synergistically killed otherwise drug-resistant cells when combined with etoposide. Athymic rats and mice carrying neuroblastoma xenografts [SH-SY5Y, SK-N-BE(2)] were treated with equimolar doses of melphalan, J1, or no drug, and effects on tumor growth and tissue morphology were analyzed. Tumor growth in vivo was significantly inhibited by J1 compared with untreated controls. Compared with melphalan, J1 more effectively inhibited the growth of mice with SH-SY5Y xenografts, was associated with higher caspase-3 activation, fewer proliferating tumor cells, and significantly decreased mean vascular density. In conclusion, the melphalan prodrug J1 is highly active in models of neuroblastoma in vitro and in vivo, encouraging further clinical development in this patient group. [Mol Cancer Ther 2007;6(9):2409–17]
Introduction
Neuroblastoma is the most common and deadly tumor of childhood often associated with therapy-resistance. For children with metastatic disease at diagnosis or certain genetic tumor features such as MYCN amplification or 1p deletion, the survival remains poor despite intensive multimodal treatment protocols (1).
The alkylating drug melphalan is routinely used in high-dose protocols for children with advanced neuroblastoma. Some decades ago, a mixture of six oligopeptides containing the meta-isomer of melphalan (m-l-sarcolysin), designated Peptichemio, was developed by Italian researchers and initial clinical trials seemed promising with significant response rates in several common diagnoses; among them, childhood neuroblastoma (2–5).
As a result of a screening of peptides, based on pharmacologic data on Peptichemio and melphalan, a highly effective drug candidate, J1 (l-melphalanyl-p-l-fluorophenylalanine ethyl ester), exhibiting higher in vitro and in vivo cytotoxicity than melphalan, was synthesized (6–8). J1 is rapidly incorporated into the cytoplasm followed by intracellular hydrolysis, which results in the release of melphalan. The enzymes responsible for the activation have been identified to be aminopeptidases (9), which may provide an attractive cancer target because the activity of several aminopeptidases is elevated in plasma and effusions from patients with cancer (10–12). A phase I trial with J1 in adult patients with advanced cancers is currently ongoing in Sweden.
There is a need for novel therapies able to bypass drug resistance of high-risk neuroblastoma, and based on previous experience with melphalan and Peptichemio in this diagnosis, it seemed adequate to investigate if J1 is active as a single agent or in combination with standard drugs in experimental models of neuroblastoma in vitro and in vivo. This is the first study showing that J1 effectively inhibits neuroblastoma cell growth in vitro and in vivo by the induction of apoptosis, being significantly more effective than melphalan and with additive or synergistic effects in combination with cytotoxic drugs routinely used for children with advanced neuroblastoma. Furthermore, this is the first report of J1 efficacy using the xenograft model including immunohistochemical examination of the tumors.
Materials and Methods
Chemicals and Reagents
The molecular structures of J1 and melphalan are shown in Fig. 1. J1 was synthesized as described previously (6) and dissolved in DMSO (Sigma-Aldrich) and further diluted with sterile water or PBS (Sigma-Aldrich). The DMSO concentration did not exceed 1% v/v in any experiment. For the in vivo studies, J1 was dissolved in N,N-dimethyl acetamide (Sigma-Aldrich) and further diluted in glucose solution (50 mg/mL; Apoteket AB). Fluorescein diacetate (Sigma-Aldrich) was dissolved in DMSO and kept frozen (−20°C) as a stock solution protected from light. Carboplatin, doxorubicin, etoposide, melphalan, and vincristine were obtained from the Swedish Pharmacy (Apoteket AB) and dissolved according to guidelines from the manufacturer and further diluted in sterile PBS (except in the xenograft studies, where melphalan was further diluted in 50 mg/mL glucose). 4-Hydroxycyclophosphamide (the active metabolite of cyclophosphamide) was a kind gift from S. Ludeman (Duke Comprehensive Cancer Center, Duke University Medical Center, Durham, NC). The aminopeptidase inhibitor bestatin (Sigma-Aldrich) was dissolved in 50% ethanol to a 4.0 mmol/L stock solution and further diluted in PBS. FAM-DEVD-FMK, supplied as part of the CaspaTag kit (Chemicon) and chloromethyl-X-rosamine (MitoTracker Red CMXRos; Molecular Probes) were dissolved in DMSO and further diluted in PBS to its final concentration. Hoechst 33342 (Sigma-Aldrich) was dissolved in water.
Human Tumor Cell Lines
Seven neuroblastoma cell lines, with different drug sensitivities, were grown in Eagle Minimal Essential Medium (SH-SY5Y; Sigma-Aldrich) or RPMI 1640 [SK-N-BE(2), SK-N-AS, SK-N-F1, SK-N-SH, SK-N-DZ, and IMR-32; Sigma-Aldrich] medium supplemented with 10% fetal bovine serum (Sigma-Aldrich) and 2 mmol/L of glutamine (Sigma-Aldrich), 100 μg/mL of streptomycin, and 100 units/mL of penicillin (Sigma-Aldrich) at 37°C humidified 5% CO2 atmosphere.
Cytotoxicity Assay
The fluorometric microculture cytotoxicity assay was used (13, 14) to investigate the effect of J1 and the combination of J1 with other cytostatic drugs in vitro. Briefly, 96- or 384-well microtiter plates (NUNC Brand Products) were pre-prepared in duplicate with drug solutions at 10 times the desired final drug concentration. Cells were seeded into the drug-prepared microtiter plates at a cell density of 0.1 × 106 cells/mL and incubated for 72 h at 37°C in a humidified 5% CO2 atmosphere. Fluorometric microculture cytotoxicity assay was done using an automated Optimized Robot for Chemical Analysis (Beckman Coulter) programmed through the software SAMI (Beckman Coulter). The plates were washed, fluorescein diacetate was added, and the fluorescence generated was measured at 485/520 nm using Fluoroscan Optima (BMG Labtech) after 50 min incubation. The resulting fluorescence is proportional to the number of intact cells in each well. A successful assay required a ratio of >5 between the signal in the control wells and the blank wells and a coefficient of variation of <30% in the control wells. Cell survival is presented as a survival index. For the combination studies, fixed concentration ratios of the drugs were used with 5-fold serial dilutions in five steps. All concentrations were tested in duplicate and the experiments were repeated thrice.
Bestatin was used to evaluate the effect of aminopeptidases on the cytotoxic effect of J1 in all seven neuroblastoma cell lines. In these analyses, cells were preincubated with 10 μmol/L of bestatin (nontoxic) for 60 min, seeded into drug-containing microtiter plates, incubated for another 30 min before the drug-containing medium was removed and fresh medium added. The activities were measured after 72 h using a fluorometric microculture cytotoxicity assay.
Apoptosis Analysis
A multiparametric high-content screening assay (15) was used for the measurement of apoptosis in the three neuroblastoma cell lines; SH-SY5Y, SK-N-AS, and SK-N-BE (2). Briefly, cells were seeded into 96-well plates (Perkin-Elmer, Inc.) and incubated with the indicated concentrations of drugs for 8 and 24 h. The FLICA probe FAM-DEVD-FMK (carboxyfluorescein-labeled fluoromethyl ketone peptide inhibitor of caspase-3; at a final concentration of 20 μmol/L) was added 1 h before the end of the drug exposure to stain activated caspase-3/7, and on the last 30 min chloromethyl-X-rosamine was included (at a final concentration of 100 nmol/L) to evaluate mitochondrial membrane potential. The plates were washed and stained with 10 μmol/L of Hoechst 33342 in 3.7% formaldehyde. Plates were analyzed using the ArrayScan high-content screening system (Cellomics®, Inc.). Images were acquired for each fluorescence channel, using suitable filters with a 20× objective, and in each well, at least 800 cells were analyzed. J1 and melphalan were tested in four different concentrations obtained by a 5-fold serial dilution. Each experiment was done thrice.
Treatment with J1 and Melphalan In vivo
Male nude rats (HsdHan: RNU-rnu; Harlan) at age 5 to 10 weeks with a weight of 150 to 275 g, and female nude mice (NMRI nu/nu; Taconic) at age 6 weeks with a weight of 18 to 25 g were used for the experiments. The animals were housed and maintained in laminar flow cabinets under specific pathogen–free conditions and given sterile water and food ad libitum. The animal experiments were approved by the regional ethics committee for animal research (N234-05 and N75-05) in accordance with the Animal Protection Law (SFS 1988:534), the Animal Protection Regulation (SFS 1988:539), and the Regulation for the Swedish National Board for Laboratory Animals (SFS 1988:541). Establishment of neuroblastoma xenografts in nude rats was done as previously described (16). For the establishment of neuroblastoma xenografts in nude mice, animals were engrafted with 30 × 106 SH-SY5Y cells (in 0.1 mL of medium) s.c. in the flank of the right hind leg using a 23-gauge needle.
Three independent experiments were carried out. One using the multidrug-resistant, MYCN-amplified, p53-mutated cell line SK-N-BE(2) in nude rats and two experiments were done using the less drug-resistant SH-SY5Y cell line, one in rats and one in mice. Initial doses were selected based on previously determined LD50 values from other strains of rats/mice, which could not separate the toxic effects of melphalan and J1 at equimolecular doses.5
Unpublished data.
Immunohistochemistry
Apoptosis, proliferation, and angiogenesis in vivo were evaluated in deparaffinized tumor sections of neuroblastoma xenografts. For identification of apoptosis, sections were incubated overnight at 4°C with a rabbit monoclonal anti–active caspase-3 antibody (1:100; R&D Systems) and detected using anti-rabbit HRP-conjugated Superpicture Polymer Kit (Zymed Laboratories, Inc.). For assessment of proliferative activity, sections were incubated with a monoclonal rabbit anti–Ki-67 antibody (1:200; Neomarkers, Inc.) overnight at 4°C and detected using an anti-rabbit HRP SuperPicture Polymer Kit (Zymed). As a control for nonspecific background staining, sections were incubated with rabbit IgG isotype control (Zymed). Biotinylated Bandeiraea simplicifolia (BS-1, L3759; Sigma-Aldrich) lectin was used for highlighting endothelial cells as described previously (17). Apoptosis and proliferation were assessed by counting the number of positively stained nuclei and the total number of tumor cells in three representative regions in three tumors from each treatment group at ×400 magnification. The results are expressed as the proportion of positively stained cells. Sections were quantified for vessel density at ×200 magnification, and fields were chosen randomly with the inclusion criteria that it had to consist of viable tissue. The results are expressed as an average number of vessels per field.
Statistical Analysis
The cytotoxic IC50 values for drugs in the cell lines in vitro were determined from log concentration-effect (survival index %) curves in GraphPad Prism (GraphPad Software, Inc.) using nonlinear regression analysis. For the in vitro results, comparison of activity between two groups was made with a two-sided t test. To test combination effects in vitro, data were analyzed using the median effect method of Chou and Talalay (18) using the software CalcuSyn Version 2 (Biosoft). Each dose-response curve (individual agents as well as combinations) were fit to a linear model using the median effect equation, allowing calculation of a median effect value D (corresponding to the IC50) and slope. Goodness-of-fit was assessed using the linear correlation coefficient, r, and r > 0.85 was required for a successful analysis. The extent of interaction between the drugs was expressed using the combination index (CI) for mutually exclusive drugs: CI = d1/D1 + d2/D2, where D1 and D2 represent the concentrations of drugs 1 and 2 alone, required to produce a certain effect, and d1 and d2 are the concentrations of drugs 1 and 2 in combination required to produce the same effect. Different CI values are obtained when solving the equation for different effect levels and a 70% effect was chosen for presentation. A CI lying on one indicates additivity; CI significantly lower was defined as synergy and CI significantly higher as antagonism. One-sample t tests were used to determine if the CIs differed from one (1.0). For the in vivo experiments, one-way ANOVA with Tukey multiple comparisons tests were used to compare the three treatment groups (GraphPad InStat). P < 0.05 was considered significant.
Results
The J1 Prodrug Is a Potent Inducer of Neuroblastoma Cell Death and Is Superior to Melphalan In vitro
The cytotoxic activity of J1, melphalan, and five cytotoxic drugs commonly used in the treatment of patients with neuroblastoma was investigated in seven neuroblastoma cell lines. In two of the neuroblastoma cell lines (SH-SY5Y and SK-N-SH), J1 was found to be the most effective drug with IC50 values of 0.0028 and 0.0051 μmol/L, respectively. In the remaining cell lines, only vincristine was more effective than J1 in inducing cell death (Fig. 2). J1 exhibited a significant increase (mean, 270-fold; range, 35- to 810-fold) in cytotoxicity compared with melphalan in all neuroblastoma cell lines investigated (t test, P < 0.001; Fig. 2).
The IC50 (with 95% confidence interval) of J1, melphalan, and five cytotoxic drugs commonly used in the treatment of neuroblastoma in seven neuroblastoma cell lines. The increase in activity of J1 compared with melphalan (defined as IC50 melphalan/IC50 J1, indicated by numbers) was significant (t test, P < 0.001) for all cell lines.
The IC50 (with 95% confidence interval) of J1, melphalan, and five cytotoxic drugs commonly used in the treatment of neuroblastoma in seven neuroblastoma cell lines. The increase in activity of J1 compared with melphalan (defined as IC50 melphalan/IC50 J1, indicated by numbers) was significant (t test, P < 0.001) for all cell lines.
To confirm that J1 acts as an enzymatic activated prodrug, as previously described (9), cells were pretreated with bestatin, a potent inhibitor of aminopeptidases (19). Pre-exposure to nontoxic concentration of bestatin resulted in a decreased activity of J1 in six of seven cell lines (t test, P < 0.05). In the IMR-32 cell line, the observed difference, however, did not reach statistical significance using this experimental setup. In general, bestatin pretreatment increased the IC50 by 5.5 times (paired t test, P = 0.0006). Bestatin did not affect neuroblastoma cell response to melphalan in any of the cell lines (t test, P > 0.05; paired t test P = 0.30).
J1 Potentiates Standard Neuroblastoma Chemotherapy In vitro
Combination chemotherapy, often supported with stem cell infusions, is considered as standard treatment in many clinical situations, and melphalan has been employed in several different protocols. Synergistic, additive, or antagonistic in vitro effects of J1 in combination with relevant chemotherapeutic drugs were evaluated in three neuroblastoma cell lines with different levels of drug resistance [SH-SY5Y, SK-N-AS, and SK-N-BE(2)]. As summarized in Table 1, which shows the CI at IC70, the addition of J1 to standard drugs induced significant synergistic or additive cytotoxic effects in all tested cell lines. Synergism was observed when J1 was combined with etoposide (all cell lines), and for carboplatin (SK-N-AS), and doxorubicin (SH-SY5Y). The other combinations were found to be additive (Table 1).
Effect of J1 in combination with chemotherapeutic drugs in neuroblastoma cells in vitro
Combination . | SK-N-AS . | . | SK-N-BE(2) . | . | SH-SY5Y . | . | |||
---|---|---|---|---|---|---|---|---|---|
. | CI at IC70 . | Effect . | CI at IC70 . | Effect . | CI at IC70 . | Effect . | |||
J1 + doxorubicin | 0.86 (0.63–1.1) | Additive | 0.88 (0.57–1.2) | Additive | 0.78 (0.64–0.92) | Synergistic | |||
J1 + carboplatin | 0.69 (0.38–0.99) | Synergistic | 1.1 (0.84–1.4) | Additive | 0.96 (0.33–1.6) | Additive | |||
J1 + cyclophosphamide | 0.82 (0.37–1.3) | Additive | 1.3 (0.58–2.0) | Additive | 1.0 (0.69–1.3) | Additive | |||
J1 + etoposide | 0.47 (0.44–0.49) | Synergistic | 0.75 (0.49–0.99) | Synergistic | 0.63 (0.25–0.99) | Synergistic | |||
J1 + vincristine | 1.4 (0.34–2.5) | Additive | 1.2 (0.46–1.9) | Additive | 0.78 (0.30–1.3) | Additive |
Combination . | SK-N-AS . | . | SK-N-BE(2) . | . | SH-SY5Y . | . | |||
---|---|---|---|---|---|---|---|---|---|
. | CI at IC70 . | Effect . | CI at IC70 . | Effect . | CI at IC70 . | Effect . | |||
J1 + doxorubicin | 0.86 (0.63–1.1) | Additive | 0.88 (0.57–1.2) | Additive | 0.78 (0.64–0.92) | Synergistic | |||
J1 + carboplatin | 0.69 (0.38–0.99) | Synergistic | 1.1 (0.84–1.4) | Additive | 0.96 (0.33–1.6) | Additive | |||
J1 + cyclophosphamide | 0.82 (0.37–1.3) | Additive | 1.3 (0.58–2.0) | Additive | 1.0 (0.69–1.3) | Additive | |||
J1 + etoposide | 0.47 (0.44–0.49) | Synergistic | 0.75 (0.49–0.99) | Synergistic | 0.63 (0.25–0.99) | Synergistic | |||
J1 + vincristine | 1.4 (0.34–2.5) | Additive | 1.2 (0.46–1.9) | Additive | 0.78 (0.30–1.3) | Additive |
NOTE: Mean of CI at IC70 with 95% confidence interval. Mutual exclusivity is assumed. Synergism and antagonism are defined as a CI mean statistically significantly lower/higher than one with one-sample t test (P < 0.05).
J1 Triggers Apoptosis in Neuroblastoma
In order to examine if JI can trigger the apoptotic machinery, mitochondrial transmembrane potential and caspase-3 activity were measured, and nuclear DNA was assessed for apoptotic fragmentation in three neuroblastoma cell lines (Fig. 3). Mitochondrial depolarization was observed 8 hours after the addition of J1 (data not shown). At 24 h post–drug addition, J1 and melphalan were both associated with caspase-3 activation and nuclear DNA fragmentation. Taken together, these observations indicate that J1 induces the apoptosis of neuroblastoma cells by activation of the intrinsic apoptotic pathway. Notably, the proapoptotic effects of J1 were significantly more potent compared with melphalan (t test, P < 0.001; Fig. 3).
Cell death characteristics for J1 and melphalan after 24-h exposure in the three neuroblastoma cell lines SK-N-AS (A), SK-N-BE(2) (B), and SH-SY5Y (C). The nuclear fragmentation/condensation and caspase-3/7 activity (measured using FAM-DEVD-FMK) are both expressed as a percentage of the untreated control and averaged for at least 800 cells per well. Points, means of three independent experiments; bars, SE.
Cell death characteristics for J1 and melphalan after 24-h exposure in the three neuroblastoma cell lines SK-N-AS (A), SK-N-BE(2) (B), and SH-SY5Y (C). The nuclear fragmentation/condensation and caspase-3/7 activity (measured using FAM-DEVD-FMK) are both expressed as a percentage of the untreated control and averaged for at least 800 cells per well. Points, means of three independent experiments; bars, SE.
J1 Significantly Inhibits the Growth of Established Neuroblastoma Xenografts
To investigate the effect of J1 and melphalan on neuroblastoma growth in vivo, nude rats and mice carrying either SK-N-BE(2) or SH-SY5Y xenografts were used. In the first experiment, nude rats carrying xenografts from the drug-resistant neuroblastoma cell line SK-N-BE(2) were treated with a single dose of 10 μmol/kg of J1 or melphalan, a dose reported to be close to 50% of LD50.6
Unpublished data, estimation in Sprague-Dawley rats.
Treatment with J1 inhibits neuroblastoma tumor growth in nude rats. A and C, tumor volume is illustrated as tumor volume index which is defined as the tumor volume at each day compared with the tumor volume at day 0. A and B, nude rats with SK-N-BE(2) xenografts treated i.v. with 10 μmol/kg of J1, melphalan, or no drug on day 0: points, mean tumor volume index; bars, SE (A); points, mean weight gain; bars, SE (B). Both drugs inhibited tumor growth compared with the untreated controls (P < 0.001). C and D, nude rats with SH-SY5Y xenografts treated i.v. with 0.50 μmol/kg of J1, melphalan, or no drug on day 0: points, mean tumor volume index; bars, SE (C); points, mean weight gain; bars, SE (D). J1 was significantly more effective in tumor growth inhibition compared with melphalan (P < 0.05). E and F, immunohistochemistry of percentage of caspase-3 (E) and Ki-67 (F) positively stained cells in tumors treated with J1 and melphalan, and untreated controls from SH-SY5Y xenografts. Three slides from three different tumors were analyzed (magnification, ×400). Columns, mean; bars, SE. J1 significantly increased caspase-3–positive cells compared with melphalan (P < 0.01) and cell proliferation was significantly inhibited after J1 treatment, but not after melphalan treatment, compared with untreated tumors (P < 0.05).
Treatment with J1 inhibits neuroblastoma tumor growth in nude rats. A and C, tumor volume is illustrated as tumor volume index which is defined as the tumor volume at each day compared with the tumor volume at day 0. A and B, nude rats with SK-N-BE(2) xenografts treated i.v. with 10 μmol/kg of J1, melphalan, or no drug on day 0: points, mean tumor volume index; bars, SE (A); points, mean weight gain; bars, SE (B). Both drugs inhibited tumor growth compared with the untreated controls (P < 0.001). C and D, nude rats with SH-SY5Y xenografts treated i.v. with 0.50 μmol/kg of J1, melphalan, or no drug on day 0: points, mean tumor volume index; bars, SE (C); points, mean weight gain; bars, SE (D). J1 was significantly more effective in tumor growth inhibition compared with melphalan (P < 0.05). E and F, immunohistochemistry of percentage of caspase-3 (E) and Ki-67 (F) positively stained cells in tumors treated with J1 and melphalan, and untreated controls from SH-SY5Y xenografts. Three slides from three different tumors were analyzed (magnification, ×400). Columns, mean; bars, SE. J1 significantly increased caspase-3–positive cells compared with melphalan (P < 0.01) and cell proliferation was significantly inhibited after J1 treatment, but not after melphalan treatment, compared with untreated tumors (P < 0.05).
In the second experiment, nude rats carrying SH-SY5Y xenografts were treated with a single dose of 0.50 μmol/kg of J1 or melphalan, a dose corresponding to ∼2.5% of LD50 (again estimated in Sprague-Dawley strain); thus, considerably lower than in the first experiment. Again, both treatments significantly inhibited tumor growth compared with untreated control tumors (ANOVA, P < 0.001; Fig. 4B). However, J1 was significantly more effective in tumor growth inhibition compared with melphalan using this lower concentration of drugs (Tukey, P < 0.05). Inhibitions were significant after 2 days for both J1 (ANOVA, P = 0.0029; Tukey, P < 0.01) and melphalan (Tukey, P < 0.05), compared with untreated tumors.
Previous in vivo studies with J1 were made with the hollow fiber method in mice only (8), showing the increased potency of J1 but a similar toxicity compared with equimolar doses of melphalan. This is the first presentation of J1's activity in a xenograft model, as well as its effect in a rat model for comparison, grafts were also studied in mice (having potentially different metabolisms and different maximum tolerated doses in previous studies). Tumor-bearing mice were treated i.v. with two doses of 0.50 μmol/kg, corresponding to ∼0.6% of LD50, of either J17
Unpublished data, estimation in specific pathogen–free Swiss mice.
Treatment with J1 inhibits neuroblastoma tumor growth in nude mice. A, tumor volume index (points, mean; bars, SE) from nude mice with SH-SY5Y xenografts treated i.v. with 0.50 μmol/kg of J1, melphalan, or no drug on days 0 and 6. Tumor growth was significantly inhibited by J1 compared with melphalan (P > 0.01). Two mice in the melphalan-treated group had to be killed before day 12 (at day 8 and 9) due to exceeding the maximal approved tumor size in the ethical approval, their volume indexes for the remaining days were set at the same level as that when they were killed. B, points, mean weight gain; bars, SE. C–E, immunohistochemistry of the percentage of caspase-3 (C) and Ki-67 (D) positively stained cells and microvessel density (E) in tumors treated with J1 or melphalan, and untreated controls. Three slides from three different tumors were analyzed (magnification, ×400 for Ki-67 and caspase-3; ×200 for vessels). Columns, mean; bars, SE. Treatment with J1 was associated with higher caspase-3 activation (P < 0.05), fewer proliferating tumor cells (P < 0.01), and significantly decreased mean vascular density (P < 0.05) compared with both the untreated controls and melphalan.
Treatment with J1 inhibits neuroblastoma tumor growth in nude mice. A, tumor volume index (points, mean; bars, SE) from nude mice with SH-SY5Y xenografts treated i.v. with 0.50 μmol/kg of J1, melphalan, or no drug on days 0 and 6. Tumor growth was significantly inhibited by J1 compared with melphalan (P > 0.01). Two mice in the melphalan-treated group had to be killed before day 12 (at day 8 and 9) due to exceeding the maximal approved tumor size in the ethical approval, their volume indexes for the remaining days were set at the same level as that when they were killed. B, points, mean weight gain; bars, SE. C–E, immunohistochemistry of the percentage of caspase-3 (C) and Ki-67 (D) positively stained cells and microvessel density (E) in tumors treated with J1 or melphalan, and untreated controls. Three slides from three different tumors were analyzed (magnification, ×400 for Ki-67 and caspase-3; ×200 for vessels). Columns, mean; bars, SE. Treatment with J1 was associated with higher caspase-3 activation (P < 0.05), fewer proliferating tumor cells (P < 0.01), and significantly decreased mean vascular density (P < 0.05) compared with both the untreated controls and melphalan.
Treatment with J1 Significantly Reduces Proliferation, Inhibits Angiogenesis, and Induces Apoptosis in Neuroblastoma In vivo
Having established that J1 seems more effective than melphalan in the treatment of established neuroblastoma xenografts, we investigated the effect of the drugs on the induction of apoptosis, tumor cell proliferation, and angiogenesis in SH-SY5Y xenografts.
A significantly increased level of cleaved caspase-3–positive cells was found in tumors from rats treated with J1 compared with untreated tumors (ANOVA, P = 0.0021; Tukey, P < 0.01; Fig. 4C). Furthermore, caspase-3 activity was significantly higher in J1-treated tumors than in xenografts from rats treated with melphalan (Tukey, P < 0.01; Fig. 4C). Cell proliferation as measured by Ki-67 was significantly inhibited after J1 treatment, but not after melphalan treatment compared with untreated tumors (ANOVA, P = 0.042; Tukey, P < 0.05 J1 versus control; P > 0.05 melphalan versus control; Fig. 4D).
A significantly elevated expression of cleaved caspase-3 was detected in SH-SY5Y xenografts from nude mice treated with J1 compared with melphalan-treated tumors (ANOVA, P = 0.017; Tukey, P < 0.05; Fig. 5B). Moreover, a significant reduction of tumor cell proliferation was also shown in J1-treated tumors compared with tumors treated with melphalan (ANOVA, P < 0.001; Tukey, P < 0.01; Fig. 5C). Because inhibition of angiogenesis could contribute to reduced cell proliferation and increased apoptosis, we also determined the effect of J1 and melphalan on mean vessel density. Treatment with J1 or melphalan induced a 49% (95% confidence interval, 27–72%; ANOVA, P < 0.001; Tukey, P < 0.001) or 20% (−0.35% to 41%; Tukey, P > 0.05) reduction in mean vessel density compared with untreated tumors. Mean vessel density was also significantly lower (36% reduction, 7.9–65%) in tumors treated with J1 compared with melphalan (Tukey, P < 0.05; Fig. 5D).
Discussion
Melphalan could prolong the survival of children with advanced neuroblastoma and is currently used in many high-dose protocols for this patient group (20). The present study shows for the first time that J1, a prodrug of melphalan, is highly active against human neuroblastoma cell lines in vitro and in vivo. The cytotoxic activity of J1 in vitro was found to be superior compared with melphalan, and statistical comparisons also favored J1 in terms of antiproliferative, proapoptotic, and antiangiogenic activity in vivo. These findings are particularly interesting in view of the demonstrated clinical activity of Peptichemio in neuroblastoma (4, 5).
Despite different levels of absolute sensitivity, the seven human neuroblastoma cell lines displayed a similar pattern of sensitivity against the tested drugs (Pearson correlation of log IC50, 0.85–0.99), reflecting the importance of a common cytopathologic origin. The in vitro activity of J1 was, on average, 270-fold that of melphalan, with a range from 35- to 810-fold. There is a clear tendency toward a greater difference in the generally more sensitive cell lines (i.e., SH-SY5Y and its parental cell line SK-N-SH) compared with the more resistant cell lines (Fig. 2). Thus, one or more drug targets specific for J1 may be more abundantly expressed in SH-SY5Y/SK-N-SH and the observation warrants further mechanistic studies. No obvious differences in IC50 of J1 between MYCN and non-MYCN–amplified cell lines could be observed. Both melphalan and J1 activated the intrinsic apoptotic pathway in neuroblastoma cells. However, a reduction in the levels of activated caspase-3/7 was observed for high concentrations of melphalan, which suggests that melphalan may induce neuroblastoma cell death by other mechanisms at higher concentrations (Fig. 3).
The major use of melphalan in neuroblastoma therapy is in high-dose regimens with stem cell support. For this purpose, melphalan has been evaluated in combination with, for example, cyclophosphamide (21), etoposide or carboplatin (22), and etoposide (23). In this work, we chose to study combinations with common neuroblastoma drugs representing different structural classes or mechanisms, and several interesting observations were made; most notably, J1 and the topoisomerase II inhibitor etoposide induced neuroblastoma cell death in a synergistic manner, the mean CI70 being significantly lower than one in all three cell lines. Notably, synergistic interactions between melphalan and topoisomerase inhibitors in vitro have been reported (24), and high-dose regimens of melphalan and etoposide have been tested clinically in patients with stage IV neuroblastoma and were found to be well-tolerated (23).
Previous in vivo studies were done in mice and included dose-finding toxicity and antitumor efficacy in the hollow fiber model. Monitored variables included behavior, weight change, blood cell analysis, autopsy, and histopathologic examinations of sternal bone marrow and the intestines. Early weight loss comparisons favored J1 over melphalan, but differences in all other variables were not significant (significant effects on blood cell count verses control for both drugs; ref. 8). Despite being simple, elegant, and robust, the hollow fiber method suffers from some disadvantages. For example, an almost complete lack of tumor-matrix interaction and no simple histopathologic or immunohistochemical evaluation of treatment results. The in vivo findings presented in this article showed that J1 is highly active, both against drug-sensitive (SH-SY5Y) and multidrug–resistant [SK-N-BE(2)] xenografts, at doses giving no observed toxicity except for a transient reduced weight gain using high doses of J1 in rats. Compared with melphalan, J1 seems to induce a more potent proapoptotic as well as the antiangiogenic activity in neuroblastoma in vivo. That J1, in addition to its direct antitumoral effects, is able to significantly reduce the number of blood vessels in neuroblastoma, is a novel finding. The metalloproteinase aminopeptidase N is highly expressed in vascular endothelial cells and has been shown to play multiple roles in angiogenesis (25). Preliminary findings in our lab show that aminopeptidase N is a target of J1,8
Wickström et al., unpublished observation.
One possible explanation for the somewhat moderate activity differences between J1 and melphalan compared with the in vitro data in the neuroblastoma xenograft studies in rodents may be related to the enzymatic differences between rodents and humans. The prodrug has two hydrolysis susceptible bonds, one peptide and one ester bond. Cleaving of the peptide bond (i.e., by peptidases) leads to melphalan, and in a previous publication, we presented evidence that this cleavage results in the increased activity of J1 compared with melphalan (8). Reports of extended esterase activity in rodents compared with humans have been published previously (27). This has been shown to be important for other prodrugs like CPT-11 (irinotecan) efficacy and toxicity in rodent experiments in vivo (28). With the rapid de-esterification of J1, some of the advantages compared with melphalan may be lost, e.g., because the de-esterified J1 is less lipophilic than J1, passive transportation into the cells may decrease. Indeed, de-esterified J1 also possesses reduced cytotoxic activity in vitro.9
Unpublished data.
In summary, these studies show significant activity of J1, a novel alkylating dipeptide and prodrug of melphalan, in neuroblastoma in vitro and in vivo, involving multiple parallel effects on tumor growth. Furthermore, additive or synergistic in vitro interactions with standard chemotherapeutics, particularly etoposide, are shown. These novel findings should encourage further preclinical studies to support clinical studies in children with neuroblastoma refractory to standard treatment.
Grant support: The Swedish Children's Cancer Foundation, the European Commission (contract no. QLK3-CT-2002-01956), and the Lions Cancer Research Fund.
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.
Acknowledgments
The work of Ninib Baryawno, Lotta Elfman, Helena Gleissman, and Agnes Rasmusson are gratefully acknowledged.