Purpose: To investigate the advantages of treatment with the SN-38–incorporating polymeric micelles NK012 over CPT-11 in combination with cisplatin [cis-dichlorodiammineplatinum (II) (CDDP)] in mice bearing a small cell lung cancer xenograft in terms of antitumor activity and toxicity, particularly intestinal toxicity.

Experimental Design: Cytotoxic effects were evaluated in human small cell lung cancer cell lines [H69, H82, and vascular endothelial growth factor (VEGF)–secreting cells (SBC-3/VEGF and its mock transfectant SBC-3/Neo)]. In vivo antitumor effects were evaluated in SBC-3/Neo–bearing and SBC-3/VEGF–bearing mice after NK012/CDDP or CPT-11/CDDP administration on days 0, 7, and 14. Drug distribution was analyzed by high-performance liquid chromatography or fluorescence microscopy, and the small intestine was pathologically examined.

Results: The in vitro growth-inhibitory effects of NK012 were 198- to 532-fold more potent than those of CPT-11. A significant difference in the relative tumor volume on day 30 was found between NK012/CDDP and CPT-11/CDDP treatments (P = 0.0058). Inflammatory changes in the small intestinal mucosa were rare in all NK012-treated mice but were commonly observed in CPT-11–treated mice. Moreover, a large amount of CPT-11 was excreted into the feces and high CPT-11 concentration was detected in the small intestinal epithelium. On the other hand, a small amount of NK012 was found in the feces and NK012 was weakly and uniformly distributed in the mucosal interstitium.

Conclusions: NK012/CDDP combination may be a promising candidate regimen against lung cancer without severe diarrhea toxicity and therefore warrants further clinical evaluation.

Translational Relevance

The SN-38–incorporating polymeric micelles NK012 has been shown to have significant antitumor activity against several cancer mouse models compared with CPT-11. The phase I study showed that patients treated with NK012 did not develop grade 3/4 diarrhea, one of the major adverse effects of CPT-11. Here, the antitumor activity of NK012/cisplatin combination was compared with that of CPT-11/cisplatin combination, one of the most active regimens against SCLC and NSCLC in the clinic. We also evaluated the pharmacologic and toxic profiles of the drug combinations, particularly in terms of diarrhea. NK012/cisplatin showed a significant potent antitumor activity against an SBC-3 xenograft compared with CPT-11/cisplatin. Moreover, inflammatory pathologic changes were rarely observed in the small intestinal mucosa of the NK012-treated mouse but were commonly observed in the CPT-11–treated mouse. NK012/cisplatin combination chemotherapy is thus a promising regimen against lung cancer without severe diarrhea toxicity and therefore warrants further clinical evaluation.

SN-38 or 7-ethyl-10-hydroxy-camptothecin is a biologically active metabolite of irinotecan hydrochloride (CPT-11) and is formed through CPT-11 conversion by carboxylesterases. SN-38 is active against various human cancers, such as colorectal, lung, and ovarian cancer (14). Although SN-38 shows up to 1,000-fold more potent cytotoxic activity against various cancer cell lines than CPT-11 in vitro (5), it has been clinically unavailable because of its water-insoluble nature, and the conversion rate from CPT-11 to SN-38 is <10% of the original CPT-11 volume in the body (6, 7).

The SN-38–incorporating polymeric micelles NK012 seem to have the advantage of passive targeting of the drug delivery system. In this passive targeting of drug delivery system, the drug accumulates in tumor tissue by using the enhanced permeability and retention effect (811). This enhanced permeability and retention effect is based on several pathologic mechanisms, which include hypervascularity, secretion of tumor vascular permeability factors stimulating extravasation of macromolecules including nanoparticles such as liposomes and micelles, and the absence of an effective lymphatic drainage of macromolecules accumulated in solid tumor tissue. Recent studies showed that NK012 has a significantly more potent antitumor activity than CPT-11 against small cell lung cancer (SCLC; ref. 12), colorectal cancer (13), renal cancer (14), pancreatic cancer (15), stomach cancer (16), and glioma (17).

It was previously reported that the SN-38/cis-dichlorodiammineplatinum (II) (CDDP) combination showed synergistic effects (18). The median survival of SCLC patients treated with the CPT-11/cisplatin (CDDP) combination was significantly longer than that of SCLC patients treated with the etoposide/CDDP combination in a randomized phase III study (P = 0.002) conducted by the Japanese Cooperative Oncology Group (19). Therefore, CPT-11/CDDP is considered to be one of the most active regimens against SCLC in Japan. A recent randomized phase III study showed that CPT-11/CDDP was equal to other platinum-based regimens, such as carboplatin plus paclitaxel, CDDP plus gemcitabine, and CDDP plus vinorelbine, in terms of response rate and overall survival in non-SCLC (NSCLC) patients (20).

One of the major clinically important toxic effects or dose-limiting factors of CPT-11 is severe late-onset diarrhea (2123). We previously showed that there was no significant difference in the kinetic character of free SN-38 in the small intestine of mice bearing the SCLC cell line SBC-3 and treated with NK012 and CPT-11 (12). Furthermore, in two independent phase I clinical trials in Japan (24) and the United States (25), nonhematologic toxicities were minimal and grade 3/4 diarrhea was absent.

In this context, we conducted this study to investigate the advantages of NK012/CDDP over CPT-11/CDDP in mice bearing a SCLC xenograft in terms of antitumor activity and toxic effects, particularly intestinal toxicity.

Drugs and cells. SN-38 and NK012 were prepared by Nippon Kayaku Co. Ltd. CPT-11 was purchased from Yakult Honsha Co. Ltd. CDDP was obtained from WC Heraeus GmbH & Co. KG.

Among the SCLC cell lines used, SBC-3 was kindly provided by Dr. I. Kimura (Okayama University, Okayama, Japan), and H69 and H82 were purchased from the American Type Culture Collection. SBC-3, H69, and H82 were maintained in RPMI 1640 supplemented with 10% fetal bovine serum (Cell Culture Technologies) and penicillin, streptomycin, and amphotericin B (100 units/mL, 100 μg/mL, and 25 μg/mL, respectively; Sigma) in a humidified atmosphere containing 5% CO2 at 37°C. Vascular endothelial growth factor (VEGF)–secreting cells, SBC-3/VEGF and its mock transfectant SBC-3/Neo, were generated from SBC-3 cells transfected with BMG-Neo-VEGF and BMG-Neo, as described (26).

In vitro study. The growth-inhibitory effects of NK012, CPT-11, SN-38, and CDDP were examined by tetrazolium salt–based proliferation assay (WST-8 assay; Wako Chemicals). One hundred microliters of a suspension of exponentially growing cells (1 × 105/mL ofSBC-3/Neo and SBC-3/VEGF or 1 × 106/mL of H69 and H82) were placed into the wells of a 96-well plate and incubated for 24 h at 37°C. Then, after medium removal, 100 μL of medium containing various concentrations of each drug were added to the wells and then incubated for 72 h at 37°C. After medium removal, 10 μL of WST-8 solution and 90 μL of medium were added to the wells followed by incubation for 1 h at 37°C. The growth-inhibitory effects of each drug were assessed spectrophotometrically (SpectraMax 190, Molecular Devices Corp.). The IC50 value was determined on the dose-response curves. The nature of interaction between NK012 and CDDP against SCLC cell lines, SBC-3/Neo, SBC-3/VEGF, H69, and H82, was evaluated by median-effect plot analyses and the combination index method of Chou and Talalay (27).

Experimental mice model. Female BALB/c nude mice (6 wk old) were purchased from SLC Japan. Mice were inoculated s.c. in the flank with 1 × 107 cells/150 μL cell suspension of SBC-3/Neo and SBC-3/VEGF cell lines.

All animal procedures were done in compliance with the guidelines for the care and use of experimental animals established by the Committee for Animal Experimentation of the National Cancer Center; these guidelines meet the ethical standards required by law and also comply with the guidelines for the use of experimental animals in Japan.

In vivo growth inhibition assay. When the tumor volume (TV) reached 1,500 mm3, mice were randomly divided into test groups consisting of five mice per group (day 0). Drugs were i.v. administered into the tail vein on days 0, 7, and 14. NK012 was given at SN-38 equivalent doses of 10 and 5 mg/kg/d, which are one third and one sixth of the maximum tolerated dose, respectively. The reference drug, CPT-11, was given at 22 and 10 mg/kg/d, which are one third and one sixth of the maximum tolerated dose, respectively. CDDP was simultaneously given on the same day at 2.5 mg/kg/d based on a previous report (28). In preliminary experiment, NK012 (5 mg/kg) plus CDDP (2.5 mg/kg) seemed to be superior to NK012 (5 mg/kg) alone in these tumors. NaCl solution (0.9%) was administered i.v. as normal control. The length (a) and width (b) of the tumor masses and body weight (BW) were measured twice a week, and TV was calculated using TV = (a × b2) / 2. Relative TV (RTV) on day n was calculated using RTV = TVn / TV0, where TVn is the TV on day n and TV0 is the TV on day 0. Relative BW (RBW) was calculated using RBW = BWn / BW0. Differences in RTV and RBW between the treatment groups on day 30 were analyzed using the unpaired t test.

Pharmacokinetic analysis by high-performance liquid chromatography. Female BALB/c nude mice (n = 3) bearing SBC-3/Neo and SBC-3/VEGF tumors (1,500 mm3) were used for drug pharmacokinetic analysis. NK012 or CPT-11 was administered at an equimolar dose of 20 or 30 mg/kg on day 0, respectively, as reported (12). CDDP was simultaneously given at 2.5 mg/kg. Mice were sacrificed 1, 6, 24, and 72 h (day 3) after administration. Plasma samples, tumors, upper small intestine, and feces were obtained and stored at −80°C until analysis.

SN-38 was extracted for each sample and reversed-phase high-performance liquid chromatography was done as reported (12).

Pathologic studies of small intestinal mucosa. CPT-11 and NK012 were injected to female BALB/c nude mice (n = 3) at the same dose schedules as those used in the treatment experiment. On day 14 after the last dosing, mice were sacrificed and parts of the small intestine were sampled at 5 cm from the pylolic part for the jejunum and 5 cm from the ileocecal junction for the ileum. Samples were fixed in 10% formalin, paraffin embedded, sectioned, and stained with H&E. Inflammation was scored by using an inflammation scale from − to ++, with − indicating absent inflammation, + showing mild inflammation predominantly infiltrated with lymphocytes, and ++ indicating active inflammation infiltrated with lymphocytes and neutrophils.

Distribution of NK012 or CPT-11 in small intestine by fluorescence microscopy. NK012 or CPT-11 was administered to female BALB/c nude mice at 20 or 30 mg/kg on day 0, respectively. Mice were sacrificed 1, 6, 24, and 72 h after drug injection, and the small intestine was excised at the middle portion and embedded in an OCT compound (Sakura Finetechnochemical Co. Ltd.) and frozen at −80°C. Tissue sections (5 μm thick) were prepared using a cryostatic microtome (Tissue-Tek Cryo3, Sakura Finetechnochemical). Frozen sections were examined under a fluorescence microscope (Biorevo, Keyence) at a 358-nm excitation wavelength and a 461-nm emission wavelength to evaluate NK012 or CPT-11 distribution in the small intestine. Because formulations containing SN-38 bound via ester bonds possess a particular fluorescence, both NK012 and CPT-11 were detected under the same fluorescence conditions.

Statistical analysis. Data were analyzed with Student's t test when groups showed equal variances (F test) or with Welch's test when they showed unequal variances (F test). P < 0.05 was considered significant. All statistical tests were two sided, and data were expressed as mean ± SD.

Cellular sensitivity of SCLC cells to NK012, CPT-11, SN-38, and CDDP. The IC50 values of NK012 for the SCLC cell lines ranged from 0.004 μmol/L (SBC-3/VEGF) to 0.041 μmol/L (H69; Table 1). The cytotoxic effects of NK012 were 198- to 532-fold higher than those of CPT-11, whereas those of NK012 were 1.10- to 2.00-fold lower than those of SN-38. These features were comparable with those reported previously (12, 13).

Table 1.

In vitro growth-inhibitory activity of SN-38, NK012, CPT-11, and CDDP in human SCLC cells

Cell lineIC50 (μmol/L)
SN-38NK012CPT-11CDDP
SBC-3/VEGF 0.00330 ± 0.00210 0.00365 ± 0.00005 1.11 ± 0.29 2.21 ± 0.36 
SBC-3/Neo 0.00872 ± 0.00063 0.0101 ± 0.0006 5.05 ± 0.08 12.8 ± 1.5 
H69 0.0205 ± 0.0195 0.0417 ± 0.0052 22.2 ± 5.9 6.23 ± 0.33 
H82 0.00716 ± 0.00079 0.00998 ± 0.00328 1.98 ± 0.55 4.08 ± 3.79 
Cell lineIC50 (μmol/L)
SN-38NK012CPT-11CDDP
SBC-3/VEGF 0.00330 ± 0.00210 0.00365 ± 0.00005 1.11 ± 0.29 2.21 ± 0.36 
SBC-3/Neo 0.00872 ± 0.00063 0.0101 ± 0.0006 5.05 ± 0.08 12.8 ± 1.5 
H69 0.0205 ± 0.0195 0.0417 ± 0.0052 22.2 ± 5.9 6.23 ± 0.33 
H82 0.00716 ± 0.00079 0.00998 ± 0.00328 1.98 ± 0.55 4.08 ± 3.79 

The molar ratios of NK012 to CDDP of 1:600 in SBC-3/VEGF, 1:120 in SBC-3/Neo, 1:150 in H69, and 1:400 in H82 were used for the drug combination studies based on the IC50 values of NK012 and CDDP (Table 1). The synergic to additive effect between NK012 and CDDP was observed in these SCLC cell lines (data not shown).

Antitumor activity of NK012/CDDP and CPT-11/CDDP against SBC-3/Neo and SBC-3/VEGF tumors. SBC-3/Neo and SBC-3/VEGF tumors treated with 5 mg/kg/d NK012 plus 2.5 mg/kg/d CDDP were significantly smaller than those treated with 10 mg/kg/d CPT-11 plus 2.5 mg/kg/d CDDP on day 30 (P = 0.0024, SBC-3/Neo; P = 0.0437, SBC-3/VEGF). Moreover, both tumors treated with 10 mg/kg/d NK012 plus 2.5 mg/kg/d CDDP were significantly smaller than those treated with 22 mg/kg/d CPT-11 plus 2.5 mg/kg/d CDDP on day 30 (P = 0.0058, SBC-3/Neo; P = 0.0478, SBC-3/VEGF; Fig. 1A and B). Although treatment-related BW loss was observed in mice treated with each drug combination, BW recovered to the normal level in each group by day 30 (Fig. 1C and D). A stronger antitumor activity against SBC-3/VEGF tumors was observed than against SBC-3/Neo tumors. The complete response rates achieved with 10 mg/kg/d NK012 plus 2.5 mg/kg/d CDDP were 100% and 0% for SBC-3/VEGF and SBC-3/Neo, respectively. These results further confirm our previous findings that a more potent antitumor effect of NK012 is observed in highly vascularized tumors (12).

Fig. 1.

Growth inhibitory effects of NK012/CDDP and CPT-11/CDDP on SBC-3/Neo and SBC-3/VEGF tumor xenografts. A and B, RTV in mice treated with NK012/CDDP or CPT-11/CDDP. SBC-3/Neo (A and C) and SBC-3/VEGF (B and D) tumors were inoculated s.c. into the flank of mice, as described in Materials and Methods. CPT-11 (10 mg/kg/d; Δ), CPT-11 (22 mg/kg/d; □), NK012 (5 mg/kg/d; ▴), or NK012 (10 mg/kg/d; ▪) combined with CDDP (2.5 mg/kg/d) were i.v. administered on days 0, 7, and 14. ○, NaCl solution (0.9%) was i.v. administered as normal control. Points, mean; bars, SD. *, P < 0.05. C and D, treatment-related BW loss occurred in mice treated with NK012/CDDP and CPT-11/CDDP. Points, mean; bars, SD.

Fig. 1.

Growth inhibitory effects of NK012/CDDP and CPT-11/CDDP on SBC-3/Neo and SBC-3/VEGF tumor xenografts. A and B, RTV in mice treated with NK012/CDDP or CPT-11/CDDP. SBC-3/Neo (A and C) and SBC-3/VEGF (B and D) tumors were inoculated s.c. into the flank of mice, as described in Materials and Methods. CPT-11 (10 mg/kg/d; Δ), CPT-11 (22 mg/kg/d; □), NK012 (5 mg/kg/d; ▴), or NK012 (10 mg/kg/d; ▪) combined with CDDP (2.5 mg/kg/d) were i.v. administered on days 0, 7, and 14. ○, NaCl solution (0.9%) was i.v. administered as normal control. Points, mean; bars, SD. *, P < 0.05. C and D, treatment-related BW loss occurred in mice treated with NK012/CDDP and CPT-11/CDDP. Points, mean; bars, SD.

Close modal

Pharmacokinetics of NK012 and CPT-11 after NK012/CDDP and CPT-11/CDDP administration in mice bearing SBC-3/Neo or SBC-3/VEGF tumors. After CPT-11/CDDP injection, the plasma concentrations of CPT-11 and SN-38 converted from CPT-11 decreased rapidly within 6 hours in a log-linear fashion (Fig. 2A). Those of NK012 (polymer-bound SN-38) and SN-38 released from NK012 decreased more gradually (Fig. 2A). As for the CPT-11 and free SN-38 concentrations in the SBC-3/Neo and SBC-3/VEGF tumors, they decreased rapidly within 6 hours, and almost no SN-38 converted from CPT-11 was detected at 24 hours in both tumors (Fig. 2B). In the case of NK012/CDDP administration, free SN-38 released from NK012 could be detected in the tumors even at 72 hours after administration (Fig. 2B). In contrast to the case of CPT-11/CDDP administration, the concentrations of free SN-38 released from NK012 were higher in the SBC-3/VEGF tumors than in the SBC-3/Neo tumors at any time point during the observation period (significant at 1 hour; P = 0.013).

Fig. 2.

Plasma, tumor, and small intestine concentrations of NK012, CPT-11, and free SN-38. Plasma (A), tumor (B), and small intestine (C) distribution of NK012, CPT-11, and free SN-38 after i.v. administration of CPT-11 (30 mg/kg) combined with CDDP (2.5 mg/kg) or NK012 (20 mg/kg) combined with CDDP (2.5 mg/kg). Left, SBC-3/Neo; right, SBC-3/VEGF. •, polymer-bound SN-38; ○, free SN-38 (polymer-unbound SN-38); Δ, SN-38 converted from CPT-11; ▴, CPT-11.

Fig. 2.

Plasma, tumor, and small intestine concentrations of NK012, CPT-11, and free SN-38. Plasma (A), tumor (B), and small intestine (C) distribution of NK012, CPT-11, and free SN-38 after i.v. administration of CPT-11 (30 mg/kg) combined with CDDP (2.5 mg/kg) or NK012 (20 mg/kg) combined with CDDP (2.5 mg/kg). Left, SBC-3/Neo; right, SBC-3/VEGF. •, polymer-bound SN-38; ○, free SN-38 (polymer-unbound SN-38); Δ, SN-38 converted from CPT-11; ▴, CPT-11.

Close modal

Free SN-38 concentrations in the small intestine after NK012/CDDP or CPT-11/CDDP administration were still detectable up to 72 hours in a similar fashion. CPT-11 concentrations 1 hour after CPT-11/CDDP administration were significantly higher than NK012 concentrations after NK012/CDDP administration (P = 0.0056, SBC-3/Neo; P = 0.017, SBC-3/VEGF; Fig. 2C).

These kinetic profiles in liver, spleen, lung, and kidney of free SN-38 after NK012/CDDP or CPT-11/CDDP administration were almost similar to those of NK012 or CPT-11 when administered as a single agent, as described (data not shown; ref. 12).

Intestinal toxicity of NK012, NK012/CDDP, CPT-11, and CPT-11/CDDP. Pathologic findings and characteristic mucosal changes are shown in Table 2 and Fig. 3. The small intestinal mucosa of mice in the CPT-11 or CPT-11/CDDP treatment group showed fibrotic changes, and active inflammation with cellular invasion, healed erosion, deformed glandular alignment, and glandular duct disappearance were also found. On the other hand, the small intestinal mucosa of mice in the NK012/CDDP treatment group showed only mild shortening and decreased number of villi or mild inflammatory cell invasion.

Table 2.

Pathologic analysis of small intestine after i.v. administration of drugs

Case no.Treatment groupSiteFibrosisInflammation
Control Jejunum − −  
 Control Ileum − −  
Control Jejunum − −  
 Control Ileum − −  
Control Jejunum − −  
 Control Ileum − −  
CPT-11 Jejunum  
 CPT-11 Ileum ++ Erosion 
CPT-11 Jejunum Edema 
 CPT-11 Ileum − −  
CPT-11 Jejunum − −  
 CPT-11 Ileum Erosion 
CDDP + CPT Jejunum  
 CDDP + CPT Ileum − −  
CDDP + CPT Jejunum  
 CDDP + CPT Ileum −  
CDDP + CPT Jejunum  
 CDDP + CPT Ileum − −  
10 NK012 Jejunum − −  
 NK012 Ileum − −  
11 NK012 Jejunum − −  
 NK012 Ileum − −  
12 NK012 Jejunum − −  
 NK012 Ileum −  
13 CDDP + NK012 Jejunum −  
 CDDP + NK012 Ileum −  
14 CDDP + NK012 Jejunum −  
 CDDP + NK012 Ileum − −  
15 CDDP + NK012 Jejunum −  
 CDDP + NK012 Ileum − −  
Case no.Treatment groupSiteFibrosisInflammation
Control Jejunum − −  
 Control Ileum − −  
Control Jejunum − −  
 Control Ileum − −  
Control Jejunum − −  
 Control Ileum − −  
CPT-11 Jejunum  
 CPT-11 Ileum ++ Erosion 
CPT-11 Jejunum Edema 
 CPT-11 Ileum − −  
CPT-11 Jejunum − −  
 CPT-11 Ileum Erosion 
CDDP + CPT Jejunum  
 CDDP + CPT Ileum − −  
CDDP + CPT Jejunum  
 CDDP + CPT Ileum −  
CDDP + CPT Jejunum  
 CDDP + CPT Ileum − −  
10 NK012 Jejunum − −  
 NK012 Ileum − −  
11 NK012 Jejunum − −  
 NK012 Ileum − −  
12 NK012 Jejunum − −  
 NK012 Ileum −  
13 CDDP + NK012 Jejunum −  
 CDDP + NK012 Ileum −  
14 CDDP + NK012 Jejunum −  
 CDDP + NK012 Ileum − −  
15 CDDP + NK012 Jejunum −  
 CDDP + NK012 Ileum − −  
Fig. 3.

Pathologic findings and characteristic mucosal changes in mouse. Jejunal and ileal mucosae from mice treated with NaCl solution (0.9%) as control, CPT-11 (22 mg/kg), CPT-11 (22 mg/kg) combined with CDDP (2.5 mg/kg), NK012 (10 mg/kg), or NK012 (10 mg/kg) combined with CDDP (2.5 mg/kg) on days 0, 7, and 14 were examined on day 28 after drug injections. The jejunal mucosa of mice in the CPT-11 treatment group showed healed erosion with fibrotic changes and lymphocytic invasion. Glandular arrangement was severely altered. Active inflammation with inflammatory cell invasion and disappearance of gland ducts were observed on the ileal mucosa in the CPT-11 treatment group. In the CPT-11/CDDP treatment group, the jejunal mucosa also showed healed erosion with scar-like fibrotic growth and mild inflammatory cell invasion into the ileal mucosa. The jejunal and ileal mucosae in the NK012 treatment group and the ileal mucosa in the NK012/CDDP treatment group were almost the same as those in the control group, that is, without inflammatory changes. The jejunal mucosa in the NK012/CDDP treatment group showed mild shortening and decreased number of villi or mild inflammatory cell invasion.

Fig. 3.

Pathologic findings and characteristic mucosal changes in mouse. Jejunal and ileal mucosae from mice treated with NaCl solution (0.9%) as control, CPT-11 (22 mg/kg), CPT-11 (22 mg/kg) combined with CDDP (2.5 mg/kg), NK012 (10 mg/kg), or NK012 (10 mg/kg) combined with CDDP (2.5 mg/kg) on days 0, 7, and 14 were examined on day 28 after drug injections. The jejunal mucosa of mice in the CPT-11 treatment group showed healed erosion with fibrotic changes and lymphocytic invasion. Glandular arrangement was severely altered. Active inflammation with inflammatory cell invasion and disappearance of gland ducts were observed on the ileal mucosa in the CPT-11 treatment group. In the CPT-11/CDDP treatment group, the jejunal mucosa also showed healed erosion with scar-like fibrotic growth and mild inflammatory cell invasion into the ileal mucosa. The jejunal and ileal mucosae in the NK012 treatment group and the ileal mucosa in the NK012/CDDP treatment group were almost the same as those in the control group, that is, without inflammatory changes. The jejunal mucosa in the NK012/CDDP treatment group showed mild shortening and decreased number of villi or mild inflammatory cell invasion.

Close modal

We next analyzed the concentrations of NK012, CPT-11, and free SN-38 in the feces. CPT-11 concentrations at 1 hour were significantly higher than NK012 concentrations (P = 0.0021) and decreased rapidly within 24 hours but remained detectable up to 72 hours. On the other hand, NK012 (polymer-bound SN-38) could be detected at a low concentration from 72 hours (Fig. 4A). To evaluate drug distribution over time, sections of the small intestine treated with NK012 or CPT-11 were examined by fluorescence microscopy. In the sections of CPT-11–treated small intestine, strong fluorescence originating from CPT-11 was detected in the epithelium of the small intestine, whereas weaker fluorescence originating from NK012 was distributed uniformly in the mucosal interstitium (Fig. 4B).

Fig. 4.

Fecal concentrations of NK012, CPT-11, and free SN-38, and NK012 or CPT-11 distribution in the small intestine. A, distribution of NK012, CPT-11, and free SN-38 after i.v. administration of CPT-11 (30 mg/kg) or NK012 (20 mg/kg). •, polymer-bound SN-38; ○, free SN-38 (polymer-unbound SN-38); Δ, SN-38 converted from CPT-11; ▴, CPT-11. B, small intestines were excised 1, 6, 24, and 72 h after i.v. administration of CPT-11 (30 mg/kg) or NK012 (20 mg/kg). Frozen sections were examined under a fluorescence microscope at a 358-nm excitation wavelength and a 461-nm emission wavelength. NK012 and CPT-11 were visualized as blue. The first or third columns are a bright-field image and the second or forth columns are a fluorescence image. Sections of small intestines were most well visualized in bright field. First, second, third, and fourth lines from the left side are images obtained 1, 6, 24, and 72 h after drug administration, respectively. CPT-11 was strongly distributed in the epithelium of the small intestine, whereas NK012 tended to be distributed weakly and uniformly in the mucosal interstitium.

Fig. 4.

Fecal concentrations of NK012, CPT-11, and free SN-38, and NK012 or CPT-11 distribution in the small intestine. A, distribution of NK012, CPT-11, and free SN-38 after i.v. administration of CPT-11 (30 mg/kg) or NK012 (20 mg/kg). •, polymer-bound SN-38; ○, free SN-38 (polymer-unbound SN-38); Δ, SN-38 converted from CPT-11; ▴, CPT-11. B, small intestines were excised 1, 6, 24, and 72 h after i.v. administration of CPT-11 (30 mg/kg) or NK012 (20 mg/kg). Frozen sections were examined under a fluorescence microscope at a 358-nm excitation wavelength and a 461-nm emission wavelength. NK012 and CPT-11 were visualized as blue. The first or third columns are a bright-field image and the second or forth columns are a fluorescence image. Sections of small intestines were most well visualized in bright field. First, second, third, and fourth lines from the left side are images obtained 1, 6, 24, and 72 h after drug administration, respectively. CPT-11 was strongly distributed in the epithelium of the small intestine, whereas NK012 tended to be distributed weakly and uniformly in the mucosal interstitium.

Close modal

Here, we compared the antitumor activity of NK012/CDDP with CPT-11/CDDP, the latter being one of the most active regimens against SCLC and NSCLC. The present data showed that when NK012/CDDP was administered, NK012 effectively accumulated in SBC-3/VEGF tumors and sufficiently exerted antitumor effects. This suggests that CDDP did not affect the permeability of tumor vessels and NK012 retention in the tumors. Hasegawa et al. (29) reported that 17 of 24 patients showed positive immunoreactivity for the VEGF protein in tumor specimens and that elevated serum VEGF levels were associated with poor outcome in SCLC. As for NSCLC, it was reported that the percentage of VEGF-positive cells was 52 ± 33% (95% confidence interval, 41-64%; median, 70%), and this value showed a positive association with high vascular grade (P = 0.008) and poor survival (P = 0.04; ref. 30). Taking all data together, NK012/CDDP may therefore be clinically effective against lung cancers, particularly those with high VEGF production.

Pathologic examinations were also conducted to evaluate changes in the small intestinal mucosa on day 14 after treatment. This is because diarrhea is one of the clinical dose-limiting toxicities of CPT-11, and epithelial apoptosis was reported as a mucosal change induced by CPT-11 (31). This pathologic change was observed on day 6 after i.p. administration of 100 mg/kg CPT-11 daily for 4 days. We found that the CPT-11–induced mucosal change was mainly fibrosis considered to be a form of recovery change from erosion. On the other hand, the small intestinal mucosa of the mice in the NK012/CDDP treatment group showed only mild shortening and decreased number of villi or mild inflammatory cell invasion. On comparison of these changes with those caused by CDDP (31), it was found that such alterations were mainly induced by CDDP rather than NK012.

A portion of SN-38 converted from CPT-11 undergoes subsequent conjugation as induced by UDP-glucuronyltransferase to form SN-38 β-glucuronide (SN-38-Glu; ref. 32). CPT-11, SN-38, and SN-38-Glu are excreted into the bile and then reach the small intestinal lumen (32, 33). SN-38-Glu is deconjugated in the cecum and colon to regenerate SN-38 through bacterial β-glucuronidase (34). In this study, CPT-11 was excreted into feces much more than NK012 and a high CPT-11 concentration was detected in the small intestinal epithelium. It is speculated that the highly excreted CPT-11 is reabsorbed in the small intestinal epithelium and converted to SN-38 to cause damage to the intestinal mucosa. On the other hand, NK012 was uniformly distributed in the mucosal interstitium at a lower concentration, which may be related to the less mucosal damage and diarrhea than those induced by CPT-11, although NK012 was observed for longer period than CPT-11. About other toxic effects including bone marrow, liver, and kidney toxicities, there was no significant difference between NK012/CDDP and CPT-11/CDDP in the present treatment schedule (data not shown).

In conclusion, NK012/CDDP showed a significantly higher antitumor activity with no severe diarrhea toxicity than CPT-11/CDDP, one of the most active regimens against SCLC and NSCLC. The present data suggest the clinical evaluation of NK012/CDDP in patients with SCLC and NSCLC.

No potential conflicts of interest were disclosed.

Grant support: Grant-in-Aid from the Third Term Comprehensive Control Research for Cancer; Ministry of Health, Labor and Welfare grant H19-025 (K. Goto, Y. Nishiwaki, and Y. Matsumura); Ministry of Education, Culture, Sports, Science and Technology Scientific Research on Priority Areas grant 17016087 (Y. Matsumura); and Japanese Foundation for Multidisciplinary Treatment of Cancer (Y. Matsumura), and the Princess Takamatsu Cancer Research Fund (07-23908).

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 N. Mie and M. Ohtsu for their technical assistance and K. Shiina and K. Abe for their secretarial assistance.

1
Argiris A, Murren JR. Advances in chemotherapy for small cell lung cancer: single-agent activity of newer agents.
Cancer J
2001
;
7
:
228
–35.
2
Bodurka DC, Levenback C, Wolf JK, et al. Phase II trial of irinotecan in patients with metastatic epithelial ovarian cancer or peritoneal cancer.
J Clin Oncol
2003
;
21
:
291
–7.
3
Cunningham D, Pyrhonen S, James RD, et al. Randomised trial of irinotecan plus supportive care versus supportive care alone after fluorouracil failure for patients with metastatic colorectal cancer.
Lancet
1998
;
352
:
1413
–8.
4
Negoro S, Masuda N, Takada Y, et al. Randomised phase III trial of irinotecan combined with cisplatin for advanced non-small-cell lung cancer.
Br J Cancer
2003
;
88
:
335
–41.
5
Mathijssen RH, van Alphen RJ, Verweij J, et al. Clinical pharmacokinetics and metabolism of irinotecan (CPT-11).
Clin Cancer Res
2001
;
7
:
2182
–94.
6
Rothenberg ML, Kuhn JG, Burris HA III, et al. Phase I and pharmacokinetic trial of weekly CPT-11.
J Clin Oncol
1993
;
11
:
2194
–204.
7
Slatter JG, Schaaf LJ, Sams JP, et al. Pharmacokinetics, metabolism, and excretion of irinotecan (CPT-11) following i.v. infusion of [(14)C]CPT-11 in cancer patients.
Drug Metab Dispos
2000
;
28
:
423
–33.
8
Dvorak HF, Nagy JA, Dvorak JT, Dvorak AM. Identification and characterization of the blood vessels of solid tumors that are leaky to circulating macromolecules.
Am J Pathol
1988
;
133
:
95
–109.
9
Maeda H, Matsumura Y. Tumoritropic and lymphotropic principles of macromolecular drugs.
Crit Rev Ther Drug Carrier Syst
1989
;
6
:
193
–210.
10
Matsumura Y, Maeda H. A new concept for macromolecular therapeutics in cancer chemotherapy: mechanism of tumoritropic accumulation of proteins and the antitumor agent smancs.
Cancer Res
1986
;
46
:
6387
–92.
11
Matsumura Y, Maruo K, Kimura M, Yamamoto T, Konno T, Maeda H. Kinin-generating cascade in advanced cancer patients and in vitro study.
Jpn J Cancer Res
1991
;
82
:
732
–41.
12
Koizumi F, Kitagawa M, Negishi T, et al. Novel SN-38-incorporating polymeric micelles, NK012, eradicate vascular endothelial growth factor-secreting bulky tumors.
Cancer Res
2006
;
66
:
10048
–56.
13
Nakajima TE, Yasunaga M, Kano Y, et al. Synergistic antitumor activity of the novel SN-38-incorporating polymeric micelles, NK012, combined with 5-fluorouracil in a mouse model of colorectal cancer, as compared with that of irinotecan plus 5-fluorouracil.
Int J Cancer
2008
;
122
:
2148
–53.
14
Sumitomo M, Koizumi F, Asano T, et al. Novel SN-38-incorporated polymeric micelle, NK012, strongly suppresses renal cancer progression.
Cancer Res
2008
;
68
:
1631
–5.
15
Saito Y, Yasunaga M, Kuroda J, Koga Y, Matsumura Y. Enhanced distribution of NK012, a polymeric micelle-encapsulated SN-38, and sustained release of SN-38 within tumors can beat a hypovascular tumor.
Cancer Sci
2008
;
99
:
1258
–64.
16
Eguchi Nakajima T, Yanagihara K, Takigahira M, et al. Antitumor effect of SN-38-releasing polymeric micelles, NK012, on spontaneous peritoneal metastases from orthotopic gastric cancer in mice compared with irinotecan.
Cancer Res
2008
;
68
:
9318
–22.
17
Kuroda J, Kuratsu J, Yasunaga M, Koga Y, Saito Y, Matsumura Y. Potent antitumor effect of SN-38-incorporating polymeric micelle, NK012, against malignant glioma.
Int J Cancer
2009
;
124
:
2505
–11.
18
Fukuda M, Nishio K, Kanzawa F, et al. Synergism between cisplatin and topoisomerase I inhibitors, NB-506 and SN-38, in human small cell lung cancer cells.
Cancer Res
1996
;
56
:
789
–93.
19
Noda K, Nishiwaki Y, Kawahara M, et al. Irinotecan plus cisplatin compared with etoposide plus cisplatin for extensive small-cell lung cancer.
N Engl J Med
2002
;
346
:
85
–91.
20
Ohe Y, Ohashi Y, Kubota K, et al. Randomized phase III study of cisplatin plus irinotecan versus carboplatin plus paclitaxel, cisplatin plus gemcitabine, and cisplatin plus vinorelbine for advanced non-small-cell lung cancer: Four-Arm Cooperative Study in Japan.
Ann Oncol
2007
;
18
:
317
–23.
21
Ohe Y, Sasaki Y, Shinkai T, et al. Phase I study and pharmacokinetics of CPT-11 with 5-day continuous infusion.
J Natl Cancer Inst
1992
;
84
:
972
–4.
22
Masuda N, Fukuoka M, Kusunoki Y, et al. CPT-11: a new derivative of camptothecin for the treatment of refractory or relapsed small-cell lung cancer.
J Clin Oncol
1992
;
10
:
1225
–9.
23
Ohno R, Okada K, Masaoka T, et al. An early phase II study of CPT-11: a new derivative of camptothecin, for the treatment of leukemia and lymphoma.
J Clin Oncol
1990
;
8
:
1907
–12.
24
Kato K, Hamaguchi T, Shirao K, et al. Interim analysis of phase I study of NK012, polymer micelle SN-38, in patients with advanced cancer [abstract 485]. Proc Am Soc Clin Oncol 2008.
25
Burris III HA, Infante JR, Spigel DR, et al. A phase I dose-escalation study of NK012 [abstract 2538]. Proc Am Soc Clin Oncol 2008.
26
Natsume T, Watanabe J, Koh Y, et al. Antitumor activity of TZT-1027 (Soblidotin) against vascular endothelial growth factor-secreting human lung cancer in vivo.
Cancer Sci
2003
;
94
:
826
–33.
27
Chou TC, Talalay P. Quantitative analysis of dose-effect relationships: the combined effects of multiple drugs or enzyme inhibitors.
Adv Enzyme Regul
1984
;
22
:
27
–55.
28
Kudoh S, Takada M, Masuda N, et al. Enhanced antitumor efficacy of a combination of CPT-11, a new derivative of camptothecin, and cisplatin against human lung tumor xenografts.
Jpn J Cancer Res
1993
;
84
:
203
–7.
29
Hasegawa Y, Takanashi S, Okudera K, et al. Vascular endothelial growth factor level as a prognostic determinant of small cell lung cancer in Japanese patients.
Intern Med
2005
;
44
:
26
–34.
30
Giatromanolaki A, Koukourakis MI, Kakolyris S, et al. Vascular endothelial growth factor, wild-type p53, and angiogenesis in early operable non-small cell lung cancer.
Clin Cancer Res
1998
;
4
:
3017
–24.
31
Ikuno N, Soda H, Watanabe M, Oka M. Irinotecan (CPT-11) and characteristic mucosal changes in the mouse ileum and cecum.
J Natl Cancer Inst
1995
;
87
:
1876
–83.
32
Atsumi R, Suzuki W, Hakusui H. Identification of the metabolites of irinotecan, a new derivative of camptothecin, in rat bile and its biliary excretion.
Xenobiotica
1991
;
21
:
1159
–69.
33
Chu XY, Kato Y, Sugiyama Y. Multiplicity of biliary excretion mechanisms for irinotecan, CPT-11, and its metabolites in rats.
Cancer Res
1997
;
57
:
1934
–8.
34
Takasuna K, Hagiwara T, Hirohashi M, et al. Involvement of β-glucuronidase in intestinal microflora in the intestinal toxicity of the antitumor camptothecin derivative irinotecan hydrochloride (CPT-11) in rats.
Cancer Res
1996
;
56
:
3752
–7.