Purpose: To investigate the autocrine/endocrine role of Id1-induced insulin-like growth factor-II (IGF-II) in esophageal cancer, and evaluate the potential of IGF-II- and IGF-type I receptor (IGF-IR)-targeted therapies.

Experimental Design: Antibody array-based screening was used to identify differentially secreted growth factors from Id1-overexpressing esophageal cancer cells. In vitro and in vivo assays were performed to confirm the induction of IGF-II by Id1, and to study the autocrine and endocrine effects of IGF-II in promoting esophageal cancer progression. Human esophageal cancer tissue microarray was analyzed for overexpression of IGF-II and its correlation with that of Id1 and phosphorylated AKT (p-AKT). The efficacy of intratumorally injected IGF-II antibody and intraperitoneally injected cixutumumab (fully human monoclonal IGF-IR antibody) was evaluated using in vivo tumor xenograft and experimental metastasis models.

Results: Id1 overexpression induced IGF-II secretion, which promoted cancer cell proliferation, survival, and invasion by activating AKT in an autocrine manner. Overexpression of IGF-II was found in 21 of 35 (60%) esophageal cancer tissues and was associated with upregulation of Id1 and p-AKT. IGF-II secreted by Id1-overexpressing esophageal cancer xenograft could instigate the growth of distant esophageal tumors, as well as promote metastasis of circulating cancer cells. Targeting IGF-II and IGF-IR had significant suppressive effects on tumor growth and metastasis in mice. Cixutumumab treatment enhanced the chemosensitivity of tumor xenografts to fluorouracil and cisplatin.

Conclusions: The Id1–IGF-II–IGF-IR–AKT signaling cascade plays an important role in esophageal cancer progression. Blockade of IGF-II/IGF-IR signaling has therapeutic potential in the management of esophageal cancer. Clin Cancer Res; 20(10); 2651–62. ©2014 AACR.

This article is featured in Highlights of This Issue, p. 2503

Translational Relevance

Esophageal cancer ranks as the sixth most frequent cause of cancer death in the world. There is an urgent need for a better understanding of the underlying mechanisms to develop more effective therapeutic strategies for this highly lethal disease. This study uncovers an autocrine/endocrine link between Id1 and the PI3K/AKT pathway involving IGF-II and IGF-IR, which promotes esophageal cancer progression and metastasis. Cixutumumab is currently investigated in clinical trials for the treatment of non–small cell lung cancer and hepatocellular carcinoma. Here, we show that systemic treatment with cixutumumab can significantly inhibit esophageal cancer growth and metastasis, as well as sensitize tumor xenografts to fluorouracil and cisplatin treatment in vivo. The present study therefore provides important preclinical data supporting the clinical application of cixutumumab systemic therapy both as a single agent and in combination with cytotoxic chemotherapeutic drugs.

Tumor-secreted growth factors can affect tumor microenvironment (1, 2), as well as stimulate the cancer cells themselves to proliferate and develop a more malignant phenotype in an autocrine manner (3–6). A variety of growth factors and cytokines exert their effects on cancer cells by activating the membrane-associated phosphoinositide 3-kinase (PI3K)/AKT signaling pathway, which regulates a wide spectrum of cellular functions, including proliferation, survival, angiogenesis, invasion, and metastasis. We previously reported that the PI3K/AKT signaling pathway can be induced by ectopic expression of Id1 (inhibitor of differentiation or DNA binding), leading to increased proliferation and survival of esophageal cancer cells (7). The Id1 protein is a helix-loop-helix protein (8) found to be overexpressed in many types of human cancer, including esophageal squamous cell carcinoma (ESCC; ref. 9). Our tissue microarray (TMA) study established that dysregulated Id1 expression is associated with tumor invasion and metastasis in ESCC (10).

The mechanism by which Id1 stimulates the PI3K/AKT pathway is not clear. In this study, using growth factor antibody array, we provided the first evidence that Id1 increases the expression and secretion of insulin-like growth factor-II (IGF-II) in a variety of human cancer types. More importantly, cancer cell-secreted IGF-II was found to mediate the effect of Id1 on the PI3K/AKT pathway. A series of in vitro and in vivo experiments were performed to study the autocrine and endocrine function of IGF-II in promoting ESCC growth and progression.

The IGF system is an attractive target for cancer therapy (11, 12). Elevated expressions of insulin-like growth factor-type I receptor (IGF-IR), and its ligands IGF-I and IGF-II, had been reported in human cancers, including ESCC, and were found to be correlated with advanced tumor stage and metastasis (13, 14), but treatment efficacy of IGF-IR blockade in ESCC remains to be evaluated. The feasibility of targeting the IGF-II-IGF-IR axis as potential treatment options for ESCC was investigated in this study in vitro, as well as in vivo using tumorigenesis and cancer metastasis mouse models. Because treatment failure and cancer recurrence may be attributed to chemoresistance during conventional chemotherapy, we also examined whether IGF-IR blockade can enhance the chemosensitivity of ESCC cells in animal models.

Cell lines and transfection

Human ESCC cell lines HKESC-3 (15), KYSE150, KYSE270, KYSE410 (16), and T.Tn (17) were used in this study. The Id1-overexpressing cell lines HKESC-3-Id1, KYSE150-Id1, and T.Tn-Id1 and their corresponding control cell lines HKESC-3-CON, KYSE150-CON, and T.Tn-CON were generated previously (18, 19). Details on reagents and plasmids used in gene overexpression and knockdown, and in generating KYSE150-Luc cell line, are given in Supplementary Materials and Methods.

Growth factor antibody array

Conditioned culture media from HKESC-3-Id1 and HKESC-3-CON cells were analyzed using a human growth factor antibody array that detects 41 growth factors (Catalog No. AAH-GF-1, RayBiotech) according to the manufacturer's instructions. The immunosignals were detected with the ECL Plus system (Amersham) and exposed to BioMax Light Film (Kodak).

In vitro ELISA, quantitative real-time PCR, Western blot analysis, MTT, TUNEL, cell migration, invasion, and soft agar assays

The concentration of human IGF-II in the culture medium and mouse serum was measured using a Non-Extraction IGF-II ELISA Kit (Diagnostic Systems Laboratories) according to the manufacturer's instruction. Anti-IGF-II (R&D Systems) was used in immunoneutralization experiments. Details of assays were described in our previous studies (19, 20). Western blot bands were quantified using ImageJ software. See Supplementary Materials and Methods for details of antibodies and real-time PCR.

In vivo tumorigenesis and metastasis experiments

Animal experiments were conducted and subcutaneous tumor volume and metastatic tumor nodules in the lungs of mice were quantified as previously described (19, 20). Details are provided in Supplementary Materials and Methods.

Immunohistochemistry

A TMA containing 35 cases of human ESCC in duplicated cores (Catalog no. ES802, Biomax), sections of tumor xenografts and mouse lung tissues were processed for immunohistochemistry and TUNEL (terminal deoxynucleotidyl transferase–mediated dUTP nick end labeling) assay. Staining intensity (for Id1, IGF-II, and phosphorylated AKT; p-AKT), Ki-67 proliferative index, TUNEL-positive apoptotic index, and CD31-positive microvessel density (MVD) were assessed as previously described (19, 20). See Supplementary Materials and Methods for details.

Statistical analysis

The data were expressed as the mean ± SD and compared using ANOVA. Correlations of IGF-II, Id1 and p-AKT in the TMA were assessed using Spearman rank correlation coefficient. P values < 0.05 were deemed significant. All in vitro experiments were repeated at least three times.

Cancer cell-secreted growth factors mediate the activation of the PI3K/AKT signaling pathway and the induction of cell proliferation by Id1

We investigated the involvement of autocrine mechanisms in the induction of PI3K/AKT by Id1. We found that the HKESC-3-Id1 conditioned medium (CM) induced protein expression of p-AKT and cell proliferation in the parental HKESC-3 cells; the AKT activation was attenuated when the proteins in the conditioned medium were denatured (Fig. 1A and B). These results suggest that Id1-overexpressing ESCC cells had increased secretion of growth factor(s) that served as autocrine stimulator of the PI3K/AKT pathway and cancer progression.

Figure 1.

Identification of the growth factors secreted from Id1-overexpressing cells. A, the HKESC-3 cells treated with the conditioned medium (CM) from HKESC-3-Id1 (with and without heat inactivation), and HKESC-3-CON cells for 1 hour were harvested for Western blot analysis. B, HKESC-3 cells were incubated for up to 72 hours in conditioned medium from HKESC-3-CON and HKESC-3-Id1, and their growth rates determined using MTT assay. C, growth factor antibody array was incubated with the serum-free conditioned medium from HKESC-3-Id1 and HKESC-3-CON cells. The representative IGF-II and internal control (IgG) spots showed higher concentration of IGF-II in the HKESC-3-Id1 conditioned medium. D, Western blot analysis of IGF-II in the cell lysates and conditioned medium obtained from three Id1-overexpressing ESCC cell lines and the respective vector control cell lines which had been kept in serum-free medium for 24 hours. E and F, KYSE150-CON-shCON, KYSE150-Id1-shCON, and KYSE150-Id1-shIGF-II cells were compared for the expression level of IGF-II in cell lysate (E) and the conditioned medium (F) by Western blot and ELISA, respectively. G, Western blot analysis of IGF-II in cell lysates and conditioned medium of KYSE150 cells transfected with different doses of pcDNA3-Id1 expression vector or siRNA against Id1. **, P < 0.01; *** P < 0.001 compared with control.

Figure 1.

Identification of the growth factors secreted from Id1-overexpressing cells. A, the HKESC-3 cells treated with the conditioned medium (CM) from HKESC-3-Id1 (with and without heat inactivation), and HKESC-3-CON cells for 1 hour were harvested for Western blot analysis. B, HKESC-3 cells were incubated for up to 72 hours in conditioned medium from HKESC-3-CON and HKESC-3-Id1, and their growth rates determined using MTT assay. C, growth factor antibody array was incubated with the serum-free conditioned medium from HKESC-3-Id1 and HKESC-3-CON cells. The representative IGF-II and internal control (IgG) spots showed higher concentration of IGF-II in the HKESC-3-Id1 conditioned medium. D, Western blot analysis of IGF-II in the cell lysates and conditioned medium obtained from three Id1-overexpressing ESCC cell lines and the respective vector control cell lines which had been kept in serum-free medium for 24 hours. E and F, KYSE150-CON-shCON, KYSE150-Id1-shCON, and KYSE150-Id1-shIGF-II cells were compared for the expression level of IGF-II in cell lysate (E) and the conditioned medium (F) by Western blot and ELISA, respectively. G, Western blot analysis of IGF-II in cell lysates and conditioned medium of KYSE150 cells transfected with different doses of pcDNA3-Id1 expression vector or siRNA against Id1. **, P < 0.01; *** P < 0.001 compared with control.

Close modal

Id1 induces IGF-II expression and secretion

We performed antibody array-based screening to identify growth factors that were differentially secreted by Id1-overexpressing ESCC cells. The results showed a higher concentration of IGF-II in the HKESC-3-Id1 CM than in the HKESC-3-CON CM (Fig. 1C and Supplementary Fig. S1). Western blot analysis of the cell lysates and conditioned medium showed increased IGF-II expression and secretion in three different Id1-overexpressing ESCC cell lines (Fig. 1D), and this effect was successfully abrogated by shIGF-II (Fig. 1E and F). We also performed transient transfection experiments to confirm that Id1 induced the production and secretion of IGF-II in a dose-dependent manner, and that siRNA knockdown of Id1 reduced IGF-II expression (Fig. 1G). To exclude the off-target effect of siRNA, a second RNA interference sequence was used to confirm the effects of Id1 knockdown on IGF-II expression (Supplementary Fig. S2). The induction of IGF-II by Id1 was further confirmed using a panel of human cancer cell lines, including gastric, colon, liver, and prostate cancer (Supplementary Fig. S3). Furthermore, manipulation of Id1 expression in ESCC cells via overexpression or short hairpin RNA knockdown resulted in corresponding changes in expression of IGF-II mRNA (Supplementary Fig. S4), which indicate that Id1 regulates IGF-II at mRNA level.

IGF-II expression is positively correlated with Id1 and p-AKT expressions in human esophageal tumors

Immunohistochemical results in TMA showed that among the 21 of 35 cancer specimens showing upregulation (i.e., moderate to strong immunostaining) of IGF-II, 15 and 14 cases had upregulated Id1 and p-AKT expression, respectively (Supplementary Fig. S5 and Supplementary Table S1). Spearman rank correlation coefficient analysis indicated a positive correlation between IGF-II and Id1 expression (r = 0.461, P < 0.005), and between IGF-II and p-AKT (r = 0.523, P < 0.005) in the ESCC specimens. These results suggest that IGF-II expression is associated with upregulation of Id1 and p-AKT in human esophageal cancer.

Autocrine IGF-II mediates the tumor- and metastasis-promoting functions of Id1 through activation of the PI3K/AKT pathway

To determine the significance and oncogenic role of Id1-induced IGF-II in ESCC cells, neutralizing antibody against IGF-II was added to the conditioned medium from HKESC-3-Id1 cells to block the functions of IGF-II. The results showed that this treatment not only attenuated the stimulation of the PI3K/AKT pathway (Fig. 2A), but also decreased the proliferative (Fig. 2B), migratory (Fig. 2C), and survival (Fig. 2D) capabilities of HKESC-3-Id1 cells. We also studied the effects of cancer cell-derived IGF-II on anchorage-independent growth using soft agar assay. As shown in Fig. 2E, the KYSE150 cancer cells were found to have significantly enhanced colony formation ability when suspended in a mixture of agar and KYSE150-Id1-shCON conditioned medium and plated on top of the base agar layer. Moreover, in the cell invasion assay (Fig. 2F), parental T.Tn and KYSE150 cells showed higher invasive ability when suspended in the conditioned media of corresponding Id1-overexpressing transfectants (i.e., T.Tn-Id1 and KYSE150-Id1-shCON). The effects on colony formation and cell invasion were abolished by knockdown of IGF-II in Id1-overexpressing cells using shIGF-II or neutralization of IGF-II in the conditioned medium (Fig. 2E and F). Furthermore, we found that treatment with the conditioned medium from KYSE150 cells, which had higher endogenous Id1 expression (see Supplementary Fig. S6 showing endogenous Id1 and IGF-II expression levels in ESCC cell lines), enhanced the ability of HKESC-3 cells to proliferate, migrate, and survive; the effects were abolished when endogenous Id1 expression in KYSE150 cells was inhibited by siRNA against Id1 (Supplementary Fig. S7).

Figure 2.

Effects of Id1-induced IGF-II secretion on PI3K/AKT activation and on oncogenic properties of esophageal cancer cells. A–C, HKESC-3 cells were treated with conditioned medium (CM) from HKESC-3-Id1, HKESC-3-CON, or HKESC-3-Id1 preincubated with IGF-II–neutralizing antibody for 1 hour. Western blot analysis (A), MTT assay (B), and wound-healing assay (C) showed that addition of anti-IGF-II (1 μg/mL) to the HKESC-3-Id1 conditioned medium strongly attenuated its stimulatory effects on the PI3K/AKT pathway, cell growth, and migration. D, HKESC-3 cells were exposed to various conditioned medium for 12 hours, and then treated with 50 ng/mL of TNF-α for up to 36 hours. Note that the increase in resistance to TNF-α–induced apoptosis induced by HKESC-3-Id1 conditioned medium was attenuated by neutralization of IGF-II in the conditioned medium. E and F, ESCC cells were suspended in various conditioned medium with or without the addition of 1 μg/mL IGF-II–neutralizing antibody. Soft agar (E) and chamber invasion assays (F) showed that knockdown or neutralization of IGF-II attenuated the Id1-induced invasive and anchorage-independent growth abilities. *, P < 0.05; **, P < 0.01; ***, P < 0.001 compared with empty vector control.

Figure 2.

Effects of Id1-induced IGF-II secretion on PI3K/AKT activation and on oncogenic properties of esophageal cancer cells. A–C, HKESC-3 cells were treated with conditioned medium (CM) from HKESC-3-Id1, HKESC-3-CON, or HKESC-3-Id1 preincubated with IGF-II–neutralizing antibody for 1 hour. Western blot analysis (A), MTT assay (B), and wound-healing assay (C) showed that addition of anti-IGF-II (1 μg/mL) to the HKESC-3-Id1 conditioned medium strongly attenuated its stimulatory effects on the PI3K/AKT pathway, cell growth, and migration. D, HKESC-3 cells were exposed to various conditioned medium for 12 hours, and then treated with 50 ng/mL of TNF-α for up to 36 hours. Note that the increase in resistance to TNF-α–induced apoptosis induced by HKESC-3-Id1 conditioned medium was attenuated by neutralization of IGF-II in the conditioned medium. E and F, ESCC cells were suspended in various conditioned medium with or without the addition of 1 μg/mL IGF-II–neutralizing antibody. Soft agar (E) and chamber invasion assays (F) showed that knockdown or neutralization of IGF-II attenuated the Id1-induced invasive and anchorage-independent growth abilities. *, P < 0.05; **, P < 0.01; ***, P < 0.001 compared with empty vector control.

Close modal

The autocrine function of Id1-induced IGF-II in ESCC was also investigated in vivo. As shown in Fig. 3A, KYSE150-Id1-shCON cells produced significantly larger tumors than the KYSE150-CON-shCON cells in nude mice (742.8 ± 142.9 mm3 vs. 516.5 ± 101.8 mm3 on day 35; P < 0.05), but the stimulatory effect of Id1 on tumor growth was counteracted in the IGF-II knockdown group. Comparison of the tumor xenografts indicated that the larger tumor size of the KYSE150-Id1-shCON group was due to increased cell proliferation and reduced apoptosis (Fig. 3B and Supplementary Fig. S8). Western blot analysis of the tumor xenografts showed that these changes were associated with the higher expression levels of IGF-II and p-AKT in the KYSE150-Id1-shCON tumors (Fig. 3C). Moreover, the mice bearing KYSE150-Id1-shCON xenografts had significantly higher concentration of human IGF-II in the circulation compared with the other two groups (Fig. 3D), which suggests that IGF-II may function as an endocrine growth stimulator for esophageal cancer. Our results also showed that shIGF-II successfully downregulated tumor and circulating IGF-II levels in vivo, thus abolishing the activation of the PI3K/AKT pathway by Id1 (Fig. 3C and D). We also determined the contribution of IGF-II as a mediator of Id1-induced tumor metastasis using experimental metastasis assay. KYSE150-CON-shCON, KYSE150-Id1-shCON, or KYSE150-Id1-shIGF-II cells were intravenously injected into nude mice through the tail vein. Six weeks later, numerous large metastatic nodules that showed positive immunostaining for human-specific cytokeratin-8 were found in the lungs of the KYSE150-Id1-shCON group, but not in the KYSE150-CON-shCON and KYSE150-Id1-shIGF-II groups (Fig. 3E). These results therefore support that IGF-II plays a significant role in mediating the Id1-promoted metastasis of human esophageal cancer in vivo.

Figure 3.

Effects of Id1-induced IGF-II on tumorigenicity and metastasis of esophageal cancer cells in nude mice. A, growth curves of subcutaneous tumors formed by KYSE150-CON-shCON, KYSE150-Id1-shCON or KYSE150-Id1-shIGF-II cells in nude mice (n = 8 per group). B, Ki-67 proliferation and apoptosis indices of tumor xenografts. C, Western blot analysis confirmed elevated IGF-II protein levels in the KYSE150-Id1-shCON tumor xenografts, and successful knockdown of IGF-II in the KYSE-Id1-shIGF-II xenografts. D, serum IGF-II concentration was assessed by ELISA. E, KYSE150-CON-shCON, KYSE150-Id1-shCON, or KYSE150-Id1-shIGF-II cells were intravenously injected into nude mice through the tail vein (n = 8 per group). Top, representative images of lungs harvested 6 weeks postinjection (metastatic nodules are indicated by arrows). Surface metastatic nodules in the lungs were counted, and the numbers presented separately for nodules below and above 1 mm in diameter. A human-specific anti-cytokeratin-8 (CK-8) monoclonal antibody was used to distinguish human ESCC cells from the surrounding mouse pulmonary tissue (middle). Bottom, consecutive lung sections stained with hematoxylin and eosin (H&E). *, P < 0.05; **, P < 0.01; ***, P < 0.001 compared with control group inoculated with KYSE150-CON-shCON cells.

Figure 3.

Effects of Id1-induced IGF-II on tumorigenicity and metastasis of esophageal cancer cells in nude mice. A, growth curves of subcutaneous tumors formed by KYSE150-CON-shCON, KYSE150-Id1-shCON or KYSE150-Id1-shIGF-II cells in nude mice (n = 8 per group). B, Ki-67 proliferation and apoptosis indices of tumor xenografts. C, Western blot analysis confirmed elevated IGF-II protein levels in the KYSE150-Id1-shCON tumor xenografts, and successful knockdown of IGF-II in the KYSE-Id1-shIGF-II xenografts. D, serum IGF-II concentration was assessed by ELISA. E, KYSE150-CON-shCON, KYSE150-Id1-shCON, or KYSE150-Id1-shIGF-II cells were intravenously injected into nude mice through the tail vein (n = 8 per group). Top, representative images of lungs harvested 6 weeks postinjection (metastatic nodules are indicated by arrows). Surface metastatic nodules in the lungs were counted, and the numbers presented separately for nodules below and above 1 mm in diameter. A human-specific anti-cytokeratin-8 (CK-8) monoclonal antibody was used to distinguish human ESCC cells from the surrounding mouse pulmonary tissue (middle). Bottom, consecutive lung sections stained with hematoxylin and eosin (H&E). *, P < 0.05; **, P < 0.01; ***, P < 0.001 compared with control group inoculated with KYSE150-CON-shCON cells.

Close modal

Tumor-secreted IGF-II instigates growth and metastasis of distant tumor cells in an endocrine manner

Because the mice bearing Id1-expressing tumor xenografts showed elevated serum IGF-II (Fig. 3D), and exposure to exogenous IGF-II-containing culture media enhanced the in vitro tumorigenic and invasive potentials of esophageal cancer cells (Fig. 2E and F), we reasoned that, in addition to autocrine stimulation, tumor-derived IGF-II may also exert systemic endocrine effects that facilitate tumor growth at a distant site. To test this hypothesis, we constructed an in vivo experimental model (Fig. 4A) similar to that used by Weinberg's team in which more aggressive “instigator” cancer cells were injected into one flank of the experimental animal and less aggressive “responder cells into the contralateral flank (21). KYSE150-CON-shCON, KYSE150-Id1-shCON, and KYSE150-Id1-shIGF-II cells were subcutaneously injected into the left flanks of different groups of nude mice to compare their ability to instigate tumor formation of parental KYSE150 cells inoculated subcutaneously into the right flank 2 weeks later (i.e., responder). As shown in Fig. 4B, the KYSE150-Id1-shCON group developed significantly larger responding tumors than the KYSE150-CON-shCON group, but knockdown of IGF-II in the instigating tumor attenuated the growth of responding tumor to a level comparable with that of the empty vector control group. We also found increased p-AKT expression (Fig. 4C), as well as significantly higher Ki-67 proliferation and lower apoptotic indices in the responding tumors of the KYSE150-Id1-shCON group compared with the KYSE150-Id1-shIGF-II and KYSE150-CON-shCON groups (Fig. 4D and Supplementary Fig. S9), suggesting that IGF-II secreted by primary tumors can exert endocrine influence to instigate the growth of less aggressive tumors at a distant site through activation of the PI3K/AKT signaling pathway (Fig. 4E).

Figure 4.

Systemic endocrine instigation of tumor growth and metastasis by Id1-induced IGF-II. A, diagram depicting experimental model and groups. B, growth curves of KYSE150 tumor xenografts (responders) in the opposite flanks of mice that harbored different types of “instigating” tumors (n = 8). C, comparison of p-AKT expression in the responder tumors by Western blot analysis. D, comparison of Ki-67 and apoptotic indices in responding tumors. E, model of endocrine instigation of distant tumor growth by Id1-induced IGF-II. F, KYSE150 cells injected intravenously into nude mice bearing KYSE150-Id1-shCON tumor xenografts had highest propensity to metastasize to the lungs (n = 8 per group). Top, gross examination of lungs (metastatic nodules are indicated by arrows). Surface metastatic nodules in the lungs were counted, and the numbers presented separately for nodules below and above 1 mm in diameter. Middle, immunohistochemical staining for metastatic human cancer cells in lung sections using anti-CK-8. Bottom, hematoxylin and eosin (H&E) staining of adjacent lung sections. *, P < 0.05; **, P < 0.01; ***, P < 0.001 compared with empty vector control group.

Figure 4.

Systemic endocrine instigation of tumor growth and metastasis by Id1-induced IGF-II. A, diagram depicting experimental model and groups. B, growth curves of KYSE150 tumor xenografts (responders) in the opposite flanks of mice that harbored different types of “instigating” tumors (n = 8). C, comparison of p-AKT expression in the responder tumors by Western blot analysis. D, comparison of Ki-67 and apoptotic indices in responding tumors. E, model of endocrine instigation of distant tumor growth by Id1-induced IGF-II. F, KYSE150 cells injected intravenously into nude mice bearing KYSE150-Id1-shCON tumor xenografts had highest propensity to metastasize to the lungs (n = 8 per group). Top, gross examination of lungs (metastatic nodules are indicated by arrows). Surface metastatic nodules in the lungs were counted, and the numbers presented separately for nodules below and above 1 mm in diameter. Middle, immunohistochemical staining for metastatic human cancer cells in lung sections using anti-CK-8. Bottom, hematoxylin and eosin (H&E) staining of adjacent lung sections. *, P < 0.05; **, P < 0.01; ***, P < 0.001 compared with empty vector control group.

Close modal

To determine whether endocrine IGF-II can facilitate the outgrowth of disseminated tumor cells, a modified experimental metastasis assay was performed in which parental KYSE150 cells were injected intravenously into the tail vein of nude mice which were inoculated 2 weeks earlier with KYSE150-CON-shCON, KYSE150-Id1-shCON, or KYSE150-Id1-shIGF-II subcutaneous xenografts. Six weeks later, we found more metastatic nodules in the lungs of the KYSE150-Id1-shCON group, than the other groups (Fig. 4F). These findings demonstrated that Id1-overexpressing tumors can facilitate metastatic colonization of disseminated cancer cells by secreting IGF-II. This aspect may have important therapeutic implications for cancer treatment.

IGF-II neutralization suppresses esophageal cancer growth in vivo

To investigate the antitumor effects of IGF-II-neutralization on esophageal cancer, nude mice were inoculated with KYSE150 cells and given intratumoral injection of IGF-II-neutralizing antibody. As shown in Fig. 5A, treatment with IGF-II–neutralizing antibody for 15 days significantly suppressed tumor growth by approximately 49% without detectable toxic effects on body weight, the lungs, liver, or kidneys (Supplementary Fig. S10A and S10B). Histologic evaluation showed that the suppressive effect of IGF-II–neutralizing antibody on the tumor growth was due to decreased cell proliferation rate and vascular supply, as well as increased apoptosis (Supplementary Fig. S10C).

Figure 5.

Effects of IGF-II–neutralizing antibody and cixutumumab on suppressing growth of human ESCC xenograft in nude mice. A, intratumoral injection with IGF-II–neutralizing antibody significantly suppressed the growth of KYSE150 tumor xenografts (n = 8 per group). B, KYSE150 (left) and KYSE270 (right) cells were injected subcutaneously into the flanks of nude mice (n = 6 per group). Treatment of the mice with 25 or 50 mg/kg cixutumumab twice a week significantly delayed the growth of the tumor xenografts. C, Ki-67 proliferation index, MVD, and apoptotic index of tumor xenografts of nude mice treated with different dosages of cixutumumab or isotype IgG. D, Western blot analysis of expression levels of p-AKT and its downstream targets in the tumor xenografts. E, immunohistochemistry confirmed that cixutumumab treatment significantly inhibited the activity of the PI3K/AKT pathway in tumor xenografts. Bars, SD; *, P < 0.05; **, P < 0.01; ***, P < 0.001 compared with isotype IgG-treated mice.

Figure 5.

Effects of IGF-II–neutralizing antibody and cixutumumab on suppressing growth of human ESCC xenograft in nude mice. A, intratumoral injection with IGF-II–neutralizing antibody significantly suppressed the growth of KYSE150 tumor xenografts (n = 8 per group). B, KYSE150 (left) and KYSE270 (right) cells were injected subcutaneously into the flanks of nude mice (n = 6 per group). Treatment of the mice with 25 or 50 mg/kg cixutumumab twice a week significantly delayed the growth of the tumor xenografts. C, Ki-67 proliferation index, MVD, and apoptotic index of tumor xenografts of nude mice treated with different dosages of cixutumumab or isotype IgG. D, Western blot analysis of expression levels of p-AKT and its downstream targets in the tumor xenografts. E, immunohistochemistry confirmed that cixutumumab treatment significantly inhibited the activity of the PI3K/AKT pathway in tumor xenografts. Bars, SD; *, P < 0.05; **, P < 0.01; ***, P < 0.001 compared with isotype IgG-treated mice.

Close modal

IGF-IR blockade inhibits esophageal cancer growth and metastasis in vivo

Because the biologic functions of IGF-II are mainly exerted through IGF-IR, we next studied whether systemic blocking of IGF-IR may inhibit tumor growth in vivo. Cixutumumab, a fully human immunoglobulin G (IgG)-1 monoclonal antibody targeting human IGF-IR (22), was injected intraperitoneally into nude mice bearing ESCC tumor xenografts. We found a significant dose-dependent reduction of tumor volume 19 days after treatment, with decreases of 66.4% and 76.6% for KYSE150 and KYSE270 tumors, respectively, in the groups receiving 50 mg/kg cixutumumab treatment (Fig. 5B). Histologic comparison of the tumor xenografts indicated that cixutumumab treatment decreased the proliferation rate and enhanced apoptosis of cancer cells, as well as reduced tumor vascular supply (Fig. 5C and Supplementary Figs. S11 and S12). Notably, Western blot and immunohistochemical analysis of tumor xenografts suggested that the inhibitory effects of cixutumumab on tumor growth were due to inactivation of the PI3K/AKT pathway (Fig. 5D and E). The elevated expression of cleaved caspase-3 also confirmed the increased apoptosis in the tumor cells of cixutumumab-treated mice (Fig. 5D). Furthermore, although cixutumumab crossreacts with mouse IGF-IR (22), cixutumumab had no obvious adverse effects on the body weight and vital organs (Supplementary Fig. S13). To evaluate the treatment efficacy of IGF-IR blockade on metastasis, nude mice were intravenously injected with luciferase-expressing ESCC cells, then treated with cixutumumab. Bioluminescent imaging and histologic examination showed marked lung metastasis in the control groups but significantly less metastatic activity in the lungs of the cixutumumab-treated groups (Fig. 6A–C). On the basis of these in vivo data, we conclude that blockade of IGF-IR with cixutumumab can significantly suppress the growth of tumor and metastasis.

Figure 6.

Effects of cixutumumab on suppressing metastasis and chemoresistance of human esophageal cancer cells. A, treatment of the mice with 25 or 50 mg/kg cixutumumab twice a week significantly reduced the extent of lung metastasis (n = 6 per group). Representative photos of bioluminescent imaging of the mice 8 weeks after cell injection were shown here. B, bioluminescent signals detected in the region of the lungs of the mice from different treatment groups were analyzed. C, representative hematoxylin and eosin (H&E)-stained lung sections from the three groups of mice. D, MTT assay comparing the viability of fluorouracil-resistant (FR) ESCC sublines with that of parental cells 4 days after treatment with 5-FU (20 μmol/L for KYSE150 cell lines, 10 μmol/L for KYSE410 cell lines, and 5 μmol/L for KYSE270 cell lines). E, FR cells were treated with 5-FU, alone or in combination with cixutumumab (100 μg/mL), and cell viability compared using MTT assay. F, nude mice xenografted with human ESCC cells KYSE150 (left) and KYSE270 (right) were treated with cixutumumab (25 mg/kg), 5-FU (20 mg/kg), cisplatin (2 mg/kg), cixutumumab plus 5-FU, or cixutumumab plus cisplatin twice weekly (n = 6 per group). Bars, SD; *, P < 0.05; **, P < 0.01; ***, P < 0.001 compared with control.

Figure 6.

Effects of cixutumumab on suppressing metastasis and chemoresistance of human esophageal cancer cells. A, treatment of the mice with 25 or 50 mg/kg cixutumumab twice a week significantly reduced the extent of lung metastasis (n = 6 per group). Representative photos of bioluminescent imaging of the mice 8 weeks after cell injection were shown here. B, bioluminescent signals detected in the region of the lungs of the mice from different treatment groups were analyzed. C, representative hematoxylin and eosin (H&E)-stained lung sections from the three groups of mice. D, MTT assay comparing the viability of fluorouracil-resistant (FR) ESCC sublines with that of parental cells 4 days after treatment with 5-FU (20 μmol/L for KYSE150 cell lines, 10 μmol/L for KYSE410 cell lines, and 5 μmol/L for KYSE270 cell lines). E, FR cells were treated with 5-FU, alone or in combination with cixutumumab (100 μg/mL), and cell viability compared using MTT assay. F, nude mice xenografted with human ESCC cells KYSE150 (left) and KYSE270 (right) were treated with cixutumumab (25 mg/kg), 5-FU (20 mg/kg), cisplatin (2 mg/kg), cixutumumab plus 5-FU, or cixutumumab plus cisplatin twice weekly (n = 6 per group). Bars, SD; *, P < 0.05; **, P < 0.01; ***, P < 0.001 compared with control.

Close modal

Cixutumumab reverts acquired chemoresistance in fluorouracil-resistant ESCC sublines and enhances the chemosensitivity of ESCC cells in vivo

Development of drug resistance often occurs in patients treated with conventional chemotherapeutic reagents. To mimic the process by which patients acquire chemoresistance, fluorouracil (5-FU)-resistant ESCC sublines were established by treating cells with increasing doses of 5-FU (up to 40 μmol/L) for over a year, and were highly resistant to 5-FU treatment (Fig. 6D). Our results showed that the fluorouracil-resistant cells had higher expression of thymidylate synthase than the parental cells (Supplementary Fig. S14A), and that cixutumumab treatment significantly reduced the expressions of thymidylate synthase and p-AKT in the fluorouracil-resistant cells (Supplementary Fig. S14B). Cixutumumab alone could moderately inhibit the proliferation of the fluorouracil-resistant cells, and that the acquired chemoresistance in fluorouracil-resistant cells was significantly abrogated by a combination of cixutumumab and 5-FU treatment (Fig. 6E). We then proceeded to investigate the therapeutic potential of cixutumumab in combination with conventional cytotoxic chemotherapeutic drugs in vivo. Figure 6F shows that cixutumumab markedly augmented the sensitivity of ESCC tumors to 5-FU and cisplatin.

In this study, we provide the first evidence that Id1 induces IGF-II in a variety of cancer cells, and that tumor-secreted IGF-II not only promotes local tumor growth through autocrine stimulation of the PI3K/AKT pathway, but also acts systemically to instigate the growth of distant tumors and facilitates the metastasis of circulating esophageal cancer cells in an endocrine manner. Furthermore, because our results showed that neutralization of IGF-II and IGF-IR blockade had significant antitumor effects, including enhancing chemosensitivity, and inhibiting tumor growth, and metastasis in animal models, we propose targeting IGF-II or IGF-IR as a potentially promising strategy in esophageal cancer therapy.

Here, we have uncovered a novel mechanism in which Id1 activates the PI3K/AKT pathway through induction and secretion of autocrine IGF-II. Our finding that Id1 increases the expression and secretion of IGF-II is significant because to our knowledge, the only other growth factor reported to be induced by Id1 expression in cancer cells is VEGF (23, 24). However, VEGF is unlikely to account for the diverse tumor-promoting effects of Id1. Data from our and neutralization experiments strongly support IGF-II as a key growth factor that mediates the oncogenic functions of Id1.

IGF-II is overexpressed in a spectrum of human cancers, including colorectal, breast, ovarian, testicular, and liver cancer (25–29). An earlier immunohistochemical study showed that 50% of ESCC tumors express IGF-II which is associated with disease progression and recurrence (13). Incidentally, our TMA analysis showed a positive correlation between Id1 and IGF-II expressions in esophageal tumors. It was proposed that growth and metastasis of tumors can be governed on a systemic level by endocrine factors released by tumors at distant sites (21). By demonstrating that nude mice bearing Id1-expressing tumor xenografts had increased serum IGF-II, which exerted a systemic instigating effect on tumor growth at distant sites, we have provided new evidence to support this concept. Furthermore, our results also indicated that tumor-secreted IGF-II could promote the colonization and proliferation of blood-borne ESCC cells at secondary sites. These findings have important implications in controlling cancer progression. In previous studies, including our own, the role of Id1 in metastasis was thought to be limited to enhancing cell migration, cell invasion, epithelial to mesenchymal transition, and production of matrix metalloproteinase, which are the key initial steps of metastasis (19, 30, 31). Here, we further demonstrated that Id1 plays an important role in the final steps of the metastatic cascade by increasing circulating IGF-II.

Esophageal cancer is a highly lethal malignancy, and development of new therapeutic intervention is urgently needed. Anticancer reagents using neutralizing antibodies to target growth factor and their receptors have emerged as a new class of effective therapeutics for human cancer. Because previous studies showed that as high as 60% of ESCC tumors expresses IGF-IR (13), inhibition of the IGF signaling pathway was proposed as a potential therapeutic option for ESCC. However, very little has been reported about the efficacy of IGF-II targeting in preclinical studies or clinical trials. Our current study provides the first evidence that neutralizing IGF-II alone can significantly inhibit the growth of human ESCC tumor xenografts. A number of IGF-IR inhibitors are currently in clinical trials for the treatment of solid tumors (32). We found that cixutumumab not only had significant inhibitory effects on esophageal cancer growth and metastasis, but also markedly enhanced the sensitivity of ESCC tumor xenograft to 5-FU and cisplatin, which are the mainstay of chemotherapeutic regimens for esophageal cancer. Our preclinical data therefore highlight the therapeutic potential of cixutumumab in combination with cytotoxic drugs. Cixutumumab is currently being evaluated in combination with paclitaxel in phase II trial as second-line therapy for metastatic esophageal cancer (http://www.clinicaltrials.gov/ct2/show/NCT01142388?term=A12+esophageal&rank=1). The mechanisms by which cixutumumab increases the sensitivity of cancer cells to chemotherapeutics are not fully understood.

The fluorouracil-resistant cells used in this study showed high expression of thymidylate synthase, which is strongly associated with 5-FU chemoresistance (33). Activation of the AKT pathway in 5-FU chemoresistant ESCC cells and tumor xenografts has been reported (34, 35), and a recent study showed that AKT is upstream of thymidylate synthase in the 5-FU signaling events (36). Therefore, our data showing significant suppression of p-AKT and thymidylate synthase by cixutumumab treatment suggest that inhibiting the PI3K/AKT pathway and thymidylate synthase expression may be the mechanisms by which cixutumumab helps cancer cells overcome 5-FU resistance.

No potential conflicts of interest were disclosed.

Conception and design: B. Li, S.W. Tsao, D.L. Ludwig, A.L.M. Cheung

Development of methodology: B. Li, S.W. Tsao, K.W. Chan

Acquisition of data (provided animals, acquired and managed patients, provided facilities, etc.): B. Li, K.W. Chan, A.L.M. Cheung

Analysis and interpretation of data (e.g., statistical analysis, biostatistics, computational analysis): B. Li, K.W. Chan, Y.Y. Li, A.L.M. Cheung

Writing, review, and/or revision of the manuscript: B. Li, K.W. Chan, D.L. Ludwig, A.L.M. Cheung

Administrative, technical, or material support (i.e., reporting or organizing data, constructing databases): R. Novosyadlyy, Y.Y. Li, Q.Y. He

Study supervision: A.L.M. Cheung

The authors thank Prof. Yutaka Shimada (University of Toyama, Toyama, Japan) and DSMZ for the KYSE cell lines, Prof. G. Srivastava and Dr. Judy Yam (Department of Pathology, University of Hong Kong, Hong Kong, China) for the HKESC-3 cell line and luciferase expression vector, respectively, and Dr. Hitoshi Kawamata (Dokkyo University School of Medicine, Tochigi, Japan) for the T.Tn cell line. In vivo imaging data were acquired using equipment maintained by the University of Hong Kong Li Ka Shing Faculty of Medicine Faculty Core Facility.

This work was supported by the Research Grants Council of the Hong Kong SAR, China (GRF Project Nos. HKU 762610M and HKU 763111M) and The University of Hong Kong SRT Cancer research program and CRCG (Project Nos. 20067176143, 200807176012, 201007176305, and 201211159003).

The costs of publication of this article were defrayed in part by the payment of page charges. This article must therefore be hereby marked advertisement in accordance with 18 U.S.C. Section 1734 solely to indicate this fact.

1.
Polyak
K
,
Haviv
I
,
Campbell
IG
. 
Co-evolution of tumor cells and their microenvironment
.
Trends Genet
2009
;
25
:
30
8
.
2.
Elkabets
M
,
Gifford
AM
,
Scheel
C
,
Nilsson
B
,
Reinhardt
F
,
Bray
MA
, et al
Human tumors instigate granulin-expressing hematopoietic cells that promote malignancy by activating stromal fibroblasts in mice
.
J Clin Invest
2011
;
121
:
784
99
.
3.
Rick
FG
,
Schally
AV
,
Szalontay
L
,
Block
NL
,
Szepeshazi
K
,
Nadji
M
, et al
Antagonists of growth hormone-releasing hormone inhibit growth of androgen-independent prostate cancer through inactivation of ERK and Akt kinases
.
Proc Natl Acad Sci U S A
2012
;
109
:
1655
60
.
4.
Xie
Q
,
Bradley
R
,
Kang
L
,
Koeman
J
,
Ascierto
ML
,
Worschech
A
, et al
Hepatocyte growth factor (HGF) autocrine activation predicts sensitivity to MET inhibition in glioblastoma
.
Proc Natl Acad Sci U S A
2012
;
109
:
570
5
.
5.
Mo
W
,
Chen
J
,
Patel
A
,
Zhang
L
,
Chau
V
,
Li
Y
, et al
CXCR4/CXCL12 mediate autocrine cell- cycle progression in NF1-associated malignant peripheral nerve sheath tumors
.
Cell
2013
;
152
:
1077
90
.
6.
Chatterjee
S
,
Heukamp
LC
,
Siobal
M
,
Schottle
J
,
Wieczorek
C
,
Peifer
M
, et al
Tumor VEGF:VEGFR2 autocrine feed-forward loop triggers angiogenesis in lung cancer
.
J Clin Invest
2013
;
123
:
1732
40
.
7.
Li
B
,
Cheung
PY
,
Wang
X
,
Tsao
SW
,
Ling
MT
,
Wong
YC
, et al
Id-1 activation of PI3K/Akt/NFkappaB signaling pathway and its significance in promoting survival of esophageal cancer cells
.
Carcinogenesis
2007
;
28
:
2313
20
.
8.
Perk
J
,
Iavarone
A
,
Benezra
R
. 
Id family of helix-loop-helix proteins in cancer
.
Nat Rev Cancer
2005
;
5
:
603
14
.
9.
Wong
YC
,
Wang
X
,
Ling
MT
. 
Id-1 expression and cell survival
.
Apoptosis
2004
;
9
:
279
89
.
10.
Yuen
HF
,
Chan
YP
,
Chan
KK
,
Chu
YY
,
Wong
ML
,
Law
SY
, et al
Id-1 and Id-2 are markers for metastasis and prognosis in oesophageal squamous cell carcinoma
.
Br J Cancer
2007
;
97
:
1409
15
.
11.
Zha
J
,
Lackner
MR
. 
Targeting the insulin-like growth factor receptor-1R pathway for cancer therapy
.
Clin Cancer Res
2010
;
16
:
2512
7
.
12.
Heidegger
I
,
Pircher
A
,
Klocker
H
,
Massoner
P
. 
Targeting the insulin-like growth factor network in cancer therapy
.
Cancer Biol Ther
2011
;
11
:
701
7
.
13.
Imsumran
A
,
Adachi
Y
,
Yamamoto
H
,
Li
R
,
Wang
Y
,
Min
Y
, et al
Insulin-like growth factor-I receptor as a marker for prognosis and a therapeutic target in human esophageal squamous cell carcinoma
.
Carcinogenesis
2007
;
28
:
947
56
.
14.
Chava
S
,
Mohan
V
,
Shetty
PJ
,
Manolla
ML
,
Vaidya
S
,
Khan
IA
, et al
Immunohistochemical evaluation of p53, FHIT, and IGF-II gene expression in esophageal cancer
.
Dis Esophagus
2012
;
25
:
81
7
.
15.
Hu
YC
,
Lam
KY
,
Law
SY
,
Wan
TS
,
Ma
ES
,
Kwong
YL
, et al
Establishment, characterization, karyotyping, and comparative genomic hybridization analysis of HKESC-2 and HKESC-3: two newly established human esophageal squamous cell carcinoma cell lines
.
Cancer Genet Cytogenet
2002
;
135
:
120
7
.
16.
Shimada
Y
,
Imamura
M
,
Wagata
T
,
Yamaguchi
N
,
Tobe
T
. 
Characterization of 21 newly established esophageal cancer cell lines
.
Cancer
1992
;
69
:
277
84
.
17.
Kawamata
H
,
Furihata
T
,
Omotehara
F
,
Sakai
T
,
Horiuchi
H
,
Shinagawa
Y
, et al
Identification of genes differentially expressed in a newly isolated human metastasizing esophageal cancer cell line, T.Tn-AT1, by cDNA microarray
.
Cancer Sci
2003
;
94
:
699
706
.
18.
Hui
CM
,
Cheung
PY
,
Ling
MT
,
Tsao
SW
,
Wang
X
,
Wong
YC
, et al
Id-1 promotes proliferation of p53-deficient esophageal cancer cells
.
Int J Cancer
2006
;
119
:
508
14
.
19.
Li
B
,
Tsao
SW
,
Li
YY
,
Wang
X
,
Ling
MT
,
Wong
YC
, et al
Id-1 promotes tumorigenicity and metastasis of human esophageal cancer cells through activation of PI3K/AKT signaling pathway
.
Int J Cancer
2009
;
125
:
2576
85
.
20.
Li
B
,
Li
YY
,
Tsao
SW
,
Cheung
AL
. 
Targeting NF-kappaB signaling pathway suppresses tumor growth, angiogenesis, and metastasis of human esophageal cancer
.
Mol Cancer Ther
2009
;
8
:
2635
44
.
21.
McAllister
SS
,
Gifford
AM
,
Greiner
AL
,
Kelleher
SP
,
Saelzler
MP
,
Ince
TA
, et al
Systemic endocrine instigation of indolent tumor growth requires osteopontin
.
Cell
2008
;
133
:
994
1005
.
22.
Rowinsky
EK
,
Youssoufian
H
,
Tonra
JR
,
Solomon
P
,
Burtrum
D
,
Ludwig
DL
. 
IMC-A12, a human IgG1 monoclonal antibody to the insulin-like growth factor I receptor
.
Clin Cancer Res
2007
;
13
:
5549s
55s
.
23.
Ling
MT
,
Lau
TC
,
Zhou
C
,
Chua
CW
,
Kwok
WK
,
Wang
Q
, et al
Overexpression of Id-1 in prostate cancer cells promotes angiogenesis through the activation of vascular endothelial growth factor (VEGF)
.
Carcinogenesis
2005
;
26
:
1668
76
.
24.
Dong
Z
,
Wei
F
,
Zhou
C
,
Sumida
T
,
Hamakawa
H
,
Hu
Y
, et al
Silencing Id-1 inhibits lymphangiogenesis through down-regulation of VEGF-C in oral squamous cell carcinoma
.
Oral Oncol
2011
;
47
:
27
32
.
25.
Cullen
KJ
,
Smith
HS
,
Hill
S
,
Rosen
N
,
Lippman
ME
. 
Growth factor messenger RNA expression by human breast fibroblasts from benign and malignant lesions
.
Cancer Res
1991
;
51
:
4978
85
.
26.
Kim
HT
,
Choi
BH
,
Niikawa
N
,
Lee
TS
,
Chang
SI
. 
Frequent loss of imprinting of the H19 and IGF-II genes in ovarian tumors
.
Am J Med Genet
1998
;
80
:
391
5
.
27.
Nonomura
N
,
Miki
T
,
Nishimura
K
,
Kanno
N
,
Kojima
Y
,
Okuyama
A
. 
Altered imprinting of the H19 and insulin-like growth factor II genes in testicular tumors
.
J Urol
1997
;
157
:
1977
9
.
28.
Schirmacher
P
,
Held
WA
,
Yang
D
,
Chisari
FV
,
Rustum
Y
,
Rogler
CE
. 
Reactivation of insulin-like growth factor II during hepatocarcinogenesis in transgenic mice suggests a role in malignant growth
.
Cancer Res
1992
;
52
:
2549
56
.
29.
Zhang
L
,
Zhou
W
,
Velculescu
VE
,
Kern
SE
,
Hruban
RH
,
Hamilton
SR
, et al
Gene expression profiles in normal and cancer cells
.
Science
1997
;
276
:
1268
72
.
30.
Fong
S
,
Itahana
Y
,
Sumida
T
,
Singh
J
,
Coppe
JP
,
Liu
Y
, et al
Id-1 as a molecular target in therapy for breast cancer cell invasion and metastasis
.
Proc Natl Acad Sci U S A
2003
;
100
:
13543
8
.
31.
Minn
AJ
,
Gupta
GP
,
Siegel
PM
,
Bos
PD
,
Shu
W
,
Giri
DD
, et al
Genes that mediate breast cancer metastasis to lung
.
Nature
2005
;
436
:
518
24
.
32.
Scagliotti
GV
,
Novello
S
. 
The role of the insulin-like growth factor signaling pathway in non-small cell lung cancer and other solid tumors
.
Cancer Treat Rev
2012
;
38
:
292
302
.
33.
Rustum
YM
. 
Thymidylate synthase: a critical target in cancer therapy?
Front Biosci
2004
;
9
:
2467
73
.
34.
You
F
,
Aoki
K
,
Ito
Y
,
Nakashima
S
. 
AKT plays a pivotal role in the acquisition of resistance to 5-fluorouracil in human squamous carcinoma cells
.
Mol Med Rep
2009
;
2
:
609
13
.
35.
Yoshida
T
,
Miyoshi
T
,
Seike
J
,
Yamai
H
,
Takechi
H
,
Yuasa
Y
, et al
Gene expression changes in a chemoresistant model with human esophageal cancer xenografts using cDNA microarray
.
Anticancer Res
2009
;
29
:
1163
8
.
36.
Vinod
BS
,
Antony
J
,
Nair
HH
,
Puliyappadamba
VT
,
Saikia
M
,
Narayanan
SS
, et al
Mechanistic evaluation of the signaling events regulating curcumin-mediated chemosensitization of breast cancer cells to 5-fluorouracil
.
Cell Death Dis
2013
;
4
:
e505
.

Supplementary data