Vandetanib inhibits cisplatin‑resistant neuroblastoma tumor growth and invasion

  • Authors:
    • Changchun Li
    • Chao Yang
    • Guanghui Wei
  • View Affiliations

  • Published online on: February 9, 2018     https://doi.org/10.3892/or.2018.6255
  • Pages: 1757-1764
Metrics: Total Views: 0 (Spandidos Publications: | PMC Statistics: )
Total PDF Downloads: 0 (Spandidos Publications: | PMC Statistics: )


Abstract

Resistance is the major cause of cisplatin treatment failure in neuroblastoma (NB). Vandetanib is widely used in the treatment of several cancers. In the present study, we aimed to determine the potential of vandetanib in cisplatin‑resistant NB therapy. Immunohistochemistry (IHC) staining was employed to detect p‑RET and CXCR4 expression in cisplatin‑resistant or ‑sensitive NB tissues from patients. Vandetanib was added to treat selected cisplatin‑resistant SH‑SY5Y cells (SH‑SY5Y‑R); this was followed by CCK8 assay, colony formation assay, and invasion assay. Furthermore, the effect of vandetanib on subcutaneous tumor growth was investigated in mice. Our results demonstrated greater expression of p‑RET and CXCR4 in cisplatin‑resistant neuroblastomas (NBs). Vandetanib significantly inhibited SH‑SY5Y‑R cell proliferation, colony formation, and invasion, while downregulating p‑RET and CXCR4 expression. Furthermore, vandetanib was as effective as high‑dose cisplatin in impairing cisplatin‑resistant NB subcutaneous tumor growth. Notably, vandetanib caused less severe liver toxicity in mice compared with high‑dose cisplatin. In summary, this study identified Vandetanib as a potential drug for cisplatin‑resistant NB treatment.

Introduction

Neuroblastomas (NBs) are known for their unpredictable behavior; some spontaneously regress, some mature, whereas others develop into aggressive forms (1). Moreover, around 7% of all tumors observed in children are NBs, next only to leukemia and brain/central nervous system tumors (2). Notably, NB accounts for approximately 15% of childhood cancer-related mortality (2,3). Though aggressive treatment strategies such as surgery, radiation, and/or chemotherapy have improved in recent decades, the prognosis for patients with disseminated NB is grim, with a 5-year survival rate of ~30% (4,5).

Multi-agent chemotherapy, including cisplatin, rapamycin, 13-cis-retinoic acid (CRA), and vincristine, is the conventional therapy for patients with advanced stages of NB (68). However, drug resistance arises in the majority of stage IV and relapsed NB, often leading to treatment failure (9,10). Furthermore, aggressive therapy also causes severe, long-term side effects in patients, including deafness, cardiac failure, and secondary malignancies (3,11). Cisplatin is one of the frontline chemotherapeutic drugs for NB and widely used in clinical therapy (12). Unfortunately, due to acquired cisplatin resistance of NBs, the prognosis of advanced NB patients after cisplatin treatment is still poor (1315). Thus, the development of novel antitumor strategies is essential to overcome cisplatin resistance and to prevent tumor progression.

Rearranged during transfection (RET) is a receptor tyrosine kinase that is expressed in various neurons including NB. Activation of RET is correlated with poor progression of NB and associated with promoting cell proliferation and metastasis (16,17). RET is triggered by anaplastic lymphoma kinase (ALK) in NB and inhibition of RET impaired tumor growth in vivo in ALK mutated NB (16). A previous study using cell lines and primary cancer samples has also demonstrated a correlation between high C-X-C chemokine receptor type 4 (CXCR4) expression levels in NB cells and increased occurrence of bone marrow metastases (18). CXCR4 was also demonstrated to support the development of NB primary tumors (19). Thus, RET and CXCR4 are the potential therapeutic targets of NB.

Vandetanib (Caprelsa, AstraZeneca Pharmaceuticals) is a small-molecule receptor tyrosine kinase inhibitor of VEGF receptor 2 (VEGFR2), EGF receptor (EGFR), and RET tyrosine kinase activity as well as mutated RET (20,21). Vandetanib is widely used as a chemotherapeutic agent in thyroid carcinoma (2224), glioblastoma (25), non-small cell lung cancer (26), and pulmonary adenocarcinoma (27). Vandetanib has been demonstrated to inhibit NB migration and invasion by reducing CXCR4 expression (28). The combination of vandetanib with CRA was more effective in reducing tumor growth than either treatment alone in NB (29). However, whether vandetanib exhibits antitumor activity in cisplatin-resistant NB is still unclear. In the present study, we aimed to determine the potential of vandetanib in cisplatin-resistant NB therapy. The NB cell line SH-SY5Y, with a strong ability for proliferation and invasion, and which is established from a metastatic bone tumor, was used in our study.

Materials and methods

Primary NB tumors

In total, 30 diagnostic primary NB tumor samples were obtained from the Department of Pediatric Surgical Oncology, Children's Hospital of Chongqing Medical University. Research was approved by the Research Ethics Committees of Chongqing Medical University. Written informed consent was signed by the parents or guardians of the pediatric patients. The patients were classified as cisplatin-resistant and -sensitive according to the prognosis of patients with cisplatin treatment and the expression of ERCC1 gene, a marker of cisplatin sensitivity. Before surgery, four cisplatin treatments, combined with vincristine, cyclophosphamide and etoposide, were performed. During the treatments, the tumor volume was assessed by B ultrasound examination once a month. Following surgery, the tumor tissues were collected for ERCC1 mRNA detection. The patients with reduction of tumor volume and ERCC1 negative expression were classified as cisplatin-sensitive. The patients without reduction of tumor volume and ERCC1 positive expression were classified as cisplatin-resistant.

Cell culture and treatment

The NB cell line SH-SY5Y was purchased from ATCC (Manassas, VA, USA). The cells were grown at 37°C in 5% CO2 in DMEM (Invitrogen, Carlsbad, CA, USA) supplemented with 10% heat-inactivated fetal bovine serum (EMD Millipore, Billerica, MA, USA), L-glutamine, sodium pyruvate, nonessential amino acids, and penicillin/streptomycin (Sigma-Aldrich, St. Louis, MO, USA). The SH-SY5Y cells were maintained at the initial cisplatin concentration of 10 µM (IC50). The dose of cisplatin was titrated gradually to a final concentration of 80 µM after 6 weeks. The selected cisplatin-resistant SH-SY5Y cells were named SH-SY5Y-R cells, and the cisplatin-sensitive SH-SY5Y cells were named SH-SY5Y-S cells. SH-SY5Y-R cells were established and then were maintained in DMEM medium with 10% FBS containing 80 µM cisplatin.

Cell viability assay

Cells (1,000) were plated in each well of a 96-well plate. Then cisplatin at different concentrations and vandetanib (0, 2.5, 5 and 10 µM) were added into the cell and incubated for 24–72 h. Cell viability was evaluated by Cell Counting Kit-8 (CCK8) assay (Dako; Agilent Technologies, Inc., Dallas, TX, USA). The relative cell viability was calculated as the OD 450 nm of the treated group/the OD 450 nm of the blank group. The IC50 was calculated using SPSS (version 21.0; IBM SPSS, Armonk, NY, USA) according to the guidelines published by Sebaugh (30).

Colony formation assay

Cells (1,000) were plated in each well of a 6-well plate. Then vandetanib (5 µM) was added into the cells and incubated for 7–10 days. The plate was fixed with 4% paraformaldehyde for 15 min and stained with crystal violet (Beyotime, Beijing, China) for 10 min at room temperatures. The number of colonies formed were counted and analyzed.

Invasion assay

Following dilution with DMEM medium (1:5), Matrigel was added into an 8.0-µm Transwell (BD, Franklin Lakes, NJ, USA). Then, 30 min later, 2×104 SH-SY5Y-S or SH-SY5Y-R cells were added into the upper well containing serum-free medium, with or without vandetanib (5 µM) treatment. The lower well was fixed with DMEM medium containing 10% FBS. Subsequently, 24 h later, the Transwell was fixed with 4% paraformaldehyde and stained with crystal violet (Beyotime, Beijing, China). The invaded cells were counted and analyzed.

Western blotting

Western blotting analysis was performed as previously described (31). Briefly, SH-SY5Y-R cells with or without vandetanib treatment were lysed in RIPA lysis buffer (Beyotime, Beijing, China) containing 1% protease inhibitor cocktail (EMD Millipore). Following concentration determination by BCA assay (Beyotime), 10 µg of total protein was added and separated by 10–12% sodium dodecyl sulfate-polyacrylamide gel electrophoresis (SDS-PAGE). The protein was transferred to a polyvinylidene fluoride (PVDF) membrane (EMD Millipore) and blocked with 5% non-fat milk in TBS/T buffer. The following antibodies were used: p-RET (rabbit monoclonal antibody; cat no. 3221; 1:800; Cell Signaling Technology, Inc., Danvers, MA, USA), CXCR4 (rabbit monoclonal antibody; cat no. ab124824; 1:1,000; Abcam, Cambridge, UK), and GAPDH (mouse monoclonal antibody; cat no. sc-293335; 1:5,000; Santa Cruz Biotechnology, Inc., Santa Cruz, CA, USA) was used as a loading control. The density of each band was assessed with ImageJ software (NIH, Bethesda, MA, USA).

Animal study

BALB/c-nu mice (5–6 weeks old, 18–20 g) were purchased from the Model Animal Center of the Nanjing University and housed in barrier facilities on a 12-h light/dark cycle. All experimental procedures were approved by the Institutional Animal Care and Use Committee of Chongqing Medical University. All mouse care and experiments were carried out in accordance with institutional guidelines concerning animal use and care of Chongqing Medical University. One week after receiving the mice, 5×106 SH-SY5Y-R cells were subcutaneously injected into the left dorsal flank. When the tumor reached ~100 mm3, the mice were randomly assigned to four groups (n=4/group). The mice were injected intratumorally with 100 µl PBS, 20 nmol cisplatin diluted in 100 µl PBS, 100 nmol cisplatin diluted in 100 µl PBS and 0.6 mg vandetanib diluted in 100 µl PBS every day. The tumor size was assessed every five days with calipers by the same investigators and the tumor volume was calculated using the equation (length × width2 × 0.52). On day 35 post-tumor cell injection, the animals were euthanized, and the tumors were excised, weighed, and paraffin-embedded.

Immunostaining and TUNEL assay

Immunostaining was performed as previously described (32). The following antibodies were used: Anti-PCNA antibody (mouse monoclonal antibody; cat no. sc25280; 1:100; Santa Cruz Biotechnology, Inc.), anti-p-RET antibody (rabbit monoclonal antibody; cat no. 3221; 1:200; Cell Signaling Technology, Inc.), anti-CXCR4 antibody (rabbit monoclonal antibody; cat no. ab124824; 1:200; Abcam), anti-CD31 antibody (mouse monoclonal antibody; cat no. 555444; 1:100; BD Biosciences). All specimens were evaluated using Olympus BX600 microscope and Spot Flex camera (Olympus, Tokyo, Japan). The positive and total cells in 3–5 random fields were counted and analyzed.

Apoptotic DNA fragmentation was examined using an in situ DeadEnd™ Fluorometric TUNEL System assay kit (Promega, Madison, WI, USA) according to the manufacturer's protocol. The localized green fluorescence of apoptotic cells from the fuorescein-12-dUTP was detected by fluorescence microscopy. The cell nuclei were stained with DAPI (Beyotime). The apoptotic cells in 5 random fields were counted and analyzed.

Statistical analysis

All statistical analyses were carried out using SPSS 19.0 statistical software (SPSS Inc., Chicago, IL, USA). The 2-tailed Student's t-test was used to evaluate the significance of differences between two groups of data and one-way ANOVA was used for statistics in multiple groups in all pertinent experiments. All experiments were performed 3–5 times. P<0.05 was considered to indicate a statistically significant result.

Results

High expression of p-RET and CXCR4 in cisplatin-resistant NB tissues

To determine the potential utility of vandetanib in cisplatin-resistant NB patients, IHC staining was employed to analyze p-RET and CXCR4 expression in 30 NB tissue samples, which were classified as originating from either cisplatin-sensitive or -resistant patients. As shown in Fig. 1, increased p-RET- and CXCR4-positive cells were found in the cisplatin-resistant NB tissues. This suggested that p-RET and CXCR4 may play a crucial role in maintaining the cisplatin resistance of NB tissues.

High expression of p-RET and CXCR4 in cisplatin-resistant NB cells

To further investigate the expression of p-RET and CXCR4 in cisplatin-sensitive and -resistant NB cells, cisplatin was used to treat SH-SY5Y cells, and the cisplatin-resistant cells were selected and named SH-SY5Y-R. As shown in Fig. 2A and B, the IC50 values of cisplatin for SH-SY5Y-S (cisplatin-sensitive SH-SY5Y cells) and SH-SY5Y-R were approximately 10 and 130 µM, respectively. These results were identical to a previous study conducted by our laboratory (33). A CCK8 assay demonstrated higher viability of proliferation in SH-SY5Y-R cells compared with SH-SY5Y-S cells (Fig. 2C). Furthermore, increased colony formation (Fig. 2D and E) and invading cells (Fig. 2F and G) were observed in the SH-SY5Y-R cells as determined by colony formation assay and Matrigel invasion assay in vitro, respectively. Western blotting demonstrated that the expression of p-RET and CXCR4 was significantly increased in SH-SY5Y-R cells (Fig. 2H and I). Collectively, these results indicated that cisplatin-resistant NB cells exhibited increased malignancy and invasive properties, combined with upregulation of p-RET and CXCR4 expression.

Figure 2.

High expression of p-RET and CXCR4 in cisplatin-resistant NB cells. (A and B) A Cell Counting Kit-8 assay was performed to determine cell proliferation of selected cisplatin-sensitive SH-SY5Y cells (SH-SY5Y-S) and cisplatin-resistant SH-SY5Y cells (SH-SY5Y-R) that were established in our laboratory and used in a previous study (33) under different concentrations of cisplatin treatment. Data represent the means ± SD from three independent experiments. (C) A Cell Counting Kit-8 assay was performed to determine cell proliferation of SH-SY5Y-S and SH-SY5Y-R cells. Data represent the means ± SD from three independent experiments (***P<0.001). (D) A colony formation assay was performed to determine tumorgenesis of SH-SY5Y-S and SH-SY5Y-R cells. (E) The number of colonies was counted and analyzed. Data represent the means ± SD from three independent experiments (**P<0.01). (F) A Matrigel-mediated invasion assay was performed to determine invasion of SH-SY5Y-S and SH-SY5Y-R cells. Scale bar, 50 µm. (G) The number of invaded cells in 4 random selected frames was counted and analyzed. Data represent the means ± SD from three independent experiments (**P<0.01). (H) Total cellular extracts from SH-SY5Y-S and SH-SY5Y-R cells were prepared and subjected to western blotting using antibodies against p-RET and CXCR4. GAPDH was used as a loading control. (I) The relative expression of p-RET and CXCR4 was analyzed. Data represent the means ± SD from three independent experiments (**P<0.01).

Vandetanib inhibits cisplatin-resistant NB tumorigenesis and invasion in vitro

We employed vandetanib to treat cisplatin-resistant NB cells. As shown in Fig. 3A, vandetanib efficiently reduced p-RET and CXCR4 expression in SH-SY5Y-R cells. A cell viability assay demonstrated that SH-SY5Y-R cell proliferation was significantly inhibited by vandetanib in a concentration-dependent manner (Fig. 3B). In addition, we demonstrated that the IC50 of vandetanib for SH-SY5Y-R cells was ~5 µM (Fig. 3B). Thus, 5 µM vandetanib was used in the following experiments. A colony formation assay, revealed that fewer colonies were formed by the vandetanib-treated SH-SY5Y-R (Fig. 3C and D) cells. Then, a Transwell invasion assay was performed to determine the effects of vandetanib on NB. We determined that vandetanib markedly prevented SH-SY5Y-R cell invasion (Fig. 3E and F). In summary, vandetanib may be an effective agent for cisplatin-resistant NB therapy.

Vandetanib inhibits cisplatin-resistant NB tumor growth in vivo

To further investigate whether vandetanib inhibited cisplatin-resistant NB tumor growth and enhanced sensitivity of NB to cisplatin in vivo, SH-SY5Y-R cells were injected into the flank of female wild-type (WT) BALB/c nude mice to establish a subcutaneous tumor model. When the tumor volume reached ~100 mm3, vandetanib and cisplatin were administered to the mice. As shown in Fig. 4A, treatment of mice with a high dose of cisplatin (100 nmol/day) markedly reduced tumor volume (Fig. 4B) and weight (Fig. 4C) by 65.7 and 65.4%, respectively. However, a low-dose of cisplatin had no observed inhibitory effect on tumor volume (Fig. 4B) or weight (Fig. 4C). In contrast, injection of vandetanib alone significantly reduced tumor volume (Fig. 4B) and weight (Fig. 4C) by 70.8 and 71.8%, respectively. IHC staining demonstrated a reduction in p-RET and CXCR4 expression in vandetanib-treated NB tumors (Fig. 4D and E). The aforementioned data demonstrated that vandetanib may be an effective agent for cisplatin-resistant NB therapy.

Vandetanib exerts low toxicity in cisplatin-resistant NB treatment

PCNA staining and TUNEL assay were performed to detect the proliferative and apoptotic cells in NB tissues. Less proliferative cells were revealed in both high-dose cisplatin-treated and vandetanib-treated tumors (Fig. 5A). Additionally, more apoptotic cells were revealed in the aforementioned tumors (Fig. 5B). Furthermore, decreased angiogenesis was observed in the vandetanib-treated tumors than in those exposed to cisplatin alone (Fig. 5C). Notably, severe liver toxicity occurred in the high-dose cisplatin treatment group, in contrast to the low dose cisplatin and control groups (Fig. 5D). No liver toxicity was observed in the vandetanib treatment group (Fig. 5D). The aforementioned data demonstrated that vandetanib exhibited low toxicity in cisplatin-resistant NB treatment.

Discussion

New treatment strategies are clearly needed for children with recurrent or refractory NBs. In the present study we demonstrated greater expression of p-RET and CXCR4 in cisplatin-resistant NB tumors compared with cisplatin-sensitive tumors. Vandetanib rapidly inhibited cisplatin-resistant NB cell proliferation, tumorigenesis, and invasion. Vandetanib alone induced a significant reduction in cisplatin-resistant NB tumor growth in vivo in a xenograft mouse model. While high-dose cisplatin treatment yielded similar results, it caused severe liver toxicity in mice.

Cisplatin is one of the frontline chemotherapeutic drugs for NB and widely used in clinical therapy (12), however the use of cisplatin is limited due to the therapy resistance (1315). In our study, we determined that cisplatin-resistant cells possessed more aggressive characteristics. Furthermore, we determined that p-RET and CXCR4 expression was significantly upregulated in cisplatin-resistant NB cells and tumor tissues of patients. This indicated that p-RET and CXCR4 upregulation may be an adaptation to cisplatin treatment and could play a crucial role in NB cisplatin resistance. Furthermore, treatment of chemosensitive NB cells with cisplatin reversibly increased EGFR expression, whereas cisplatin-resistant cells revealed enhanced EGFR expression independent of the presence of cisplatin (34). Inhibition of EGFR, using gefitinib, revealed minor chemosensitizing effects in NB (35), whereas EGFR-targeted antibodies and growth factor toxins scFv(14E1)-Pseudomonas exotoxin A (ETA) and TGF-α-ETA exerted anticancer effects in NB cell lines (34). In the present study we revealed that EGFR expression was upregulated in cisplatin-resistant SH-SY5Y cells (data not shown), but its expression was not inhibited by vandetanib at the concentration of 5 µM (data not shown). Therefore, EGFR may be another adaptation to cisplatin treatment in NB, but it was not the effector of vandetanib in the inhibition of cisplatin-resistant NB tumor progression at low concentrations.

Increasing the concentration of cisplatin is the most common strategy to offset cisplatin resistance. However, high-dose cisplatin may cause severe liver toxicity, which is the main side-effect of cisplatin therapy (36,37). In the present study we demonstrated that vandetanib was as effective as high-dose cisplatin in impairing cisplatin-resistant NB subcutaneous tumor growth in vivo. Notably, less liver toxicity was observed in the vandetanib treatment group than in the high-dose cisplatin treatment group. These results provide solid evidence, demonstrating the advantages of vandetanib in the treatment of cisplatin-resistant NB. Whether combination of vandetanib with cisplatin produces a better therapeutic effect in NB will be investigated in a future study. Different concentrations of vandetanib will be used to treat cisplatin resistance in NB after combination with different concentrations of cisplatin. The potential synergy will be analyzed according to previous models (38,39).

Previously, RET rearrangements have been reported in NB (17). Activated ALK triggered RET upregulation in mouse sympathetic ganglia at birth, as well as in murine and human NB (16). RET inhibition strongly impaired tumor growth in vivo in both MYCN/KI AlkR1279Q and MYCN/KI AlkF1178L mice (16). Inhibition of RET phosphorylation by vandetanib treatment resulted in the induction of apoptosis in the majority of NB cell lines in vitro, and inhibited tumor growth in a mouse xenograft model, via both reduction in tumor vascularity and induction of apoptosis (16). Notably, in the present study we first demonstrated that inhibition of RET phosphorylation resulted in the inhibition of proliferation, invasion, and induction of apoptosis in cisplatin-resistant NB cells. Vandetanib treatment was an efficient therapy for cisplatin-resistant NB tumor growth, inducing apoptosis and inhibiting proliferation and angiogenesis.

CXCR4 has been demonstrated to be one of the most frequently expressed chemokines, affecting tumor cell proliferation, survival, and metastasis in various cancers (40,41). In NB, CXCR4 has been proposed to be involved in the mechanisms by which cells metastasize to specific sites from the primary site (18,42). Greater expression of CXCR4 in NBs was correlated with high-stage disease and worse clinical outcome than lower expression of CXCR4 (43,44). Functional studies have demonstrated that inhibition of CXCR4 was an efficient strategy to inhibit NB cell proliferation and metastasis (4547). A previous study by Ding et al demonstrated the inhibitory role of vandetanib on NB cell migration and invasion through downregulation of CXCR4 and MMP-14 expression (28). The present study for the first time also indicated that vandetanib treatment caused a significant decrease in CXCR4 expression and cisplatin-resistant NB cell invasion. Ding et al demonstrated that the IC50 of vandetanib for SH-SY5Y cells was ~10 µM. However we demonstrated that the IC50 of vandetanib for SH-SY5Y-R cells was ~5 µM. This could be attributed to the higher expression of CXCR4 in cisplatin-resistant SH-SY5Y cells, which enhances the sensitivity of vandetanib.

In conclusion, the present study indicated that vandetanib is an efficient therapeutic agent for cisplatin-resistant NBs, inhibiting p-RET and CXCR4 expression. It identified vandetanib as a potential therapy for cisplatin-resistant NBs. In particular, the combination of vandetanib with cisplatin may represent a novel therapeutic strategy in NB patients.

Competing interests

The authors declare that they have no competing interests.

References

1 

Nakazawa A, Haga C, Ohira M, Okita H, Kamijo T and Nakagawara A: Correlation between the international neuroblastoma pathology classification and genomic signature in neuroblastoma. Cancer Sci. 106:766–771. 2015. View Article : Google Scholar : PubMed/NCBI

2 

Maris JM: Recent advances in neuroblastoma. N Engl J Med. 362:2202–2211. 2010. View Article : Google Scholar : PubMed/NCBI

3 

Oeffinger KC, Mertens AC, Sklar CA, Kawashima T, Hudson MM, Meadows AT, Friedman DL, Marina N, Hobbie W, Kadan-Lottick NS, et al: Chronic health conditions in adult survivors of childhood cancer. N Engl J Med. 355:1572–1582. 2006. View Article : Google Scholar : PubMed/NCBI

4 

Maris JM, Hogarty MD, Bagatell R and Cohn SL: Neuroblastoma. Lancet (London, England). 369:2106–2120. 2007. View Article : Google Scholar : PubMed/NCBI

5 

Tao X: Antibody therapy and neuroblastoma. N Engl J Med. 364:289–290. 2011. View Article : Google Scholar : PubMed/NCBI

6 

Barone G, Anderson J, Pearson AD, Petrie K and Chesler L: New strategies in neuroblastoma: Therapeutic targeting of MYCN and ALK. Clin Cancer Res. 19:5814–5821. 2013. View Article : Google Scholar : PubMed/NCBI

7 

Brodeur GM, Iyer R, Croucher JL, Zhuang T, Higashi M and Kolla V: Therapeutic targets for neuroblastomas. Expert Opin Ther Targets. 18:277–292. 2014. View Article : Google Scholar : PubMed/NCBI

8 

Morgenstern DA, Baruchel S and Irwin MS: Current and future strategies for relapsed neuroblastoma: Challenges on the road to precision therapy. J Pediatr Hematol Oncol. 35:337–347. 2013. View Article : Google Scholar : PubMed/NCBI

9 

Alisi A, Cho WC, Locatelli F and Fruci D: Multidrug resistance and cancer stem cells in neuroblastoma and hepatoblastoma. Int J Mol Sci. 14:24706–24725. 2013. View Article : Google Scholar : PubMed/NCBI

10 

Fruci D, Cho WC, Nobili V, Locatelli F and Alisi A: Drug transporters and multiple drug resistance in pediatric solid tumors. Curr Drug Metab. 17:308–316. 2016. View Article : Google Scholar : PubMed/NCBI

11 

Matthay KK, Reynolds CP, Seeger RC, Shimada H, Adkins ES, Haas-Kogan D, Gerbing RB, London WB and Villablanca JG: Long-term results for children with high-risk neuroblastoma treated on a randomized trial of myeloablative therapy followed by 13-cis-retinoic acid: A children's oncology group study. J Clin Oncol. 27:1007–1013. 2009. View Article : Google Scholar : PubMed/NCBI

12 

De Bernardi B, Carli M, Casale F, Corciulo P, Cordero di Montezemolo L, De Laurentis C, Bagnulo S, Brisigotti M, Marchese N, Garaventa A, et al: Standard-dose and high-dose peptichemio and cisplatin in children with disseminated poor-risk neuroblastoma: Two studies by the Italian Cooperative Group for Neuroblastoma. J Clin Oncol. 10:1870–1878. 1992. View Article : Google Scholar : PubMed/NCBI

13 

Landier W, Knight K, Wong FL, Lee J, Thomas O, Kim H, Kreissman SG, Schmidt ML, Chen L, London WB, et al: Ototoxicity in children with high-risk neuroblastoma: Prevalence, risk factors, and concordance of grading scales-a report from the Children's Oncology Group. J Clin Oncol. 32:527–534. 2014. View Article : Google Scholar : PubMed/NCBI

14 

Vella S, Penna I, Longo L, Pioggia G, Garbati P, Florio T, Rossi F and Pagano A: Perhexiline maleate enhances antitumor efficacy of cisplatin in neuroblastoma by inducing over-expression of NDM29 ncRNA. Sci Rep. 5:181442015. View Article : Google Scholar : PubMed/NCBI

15 

Ryan J, Tivnan A, Fay J, Bryan K, Meehan M, Creevey L, Lynch J, Bray IM, O'Meara A, Tracey L, et al: MicroRNA-204 increases sensitivity of neuroblastoma cells to cisplatin and is associated with a favourable clinical outcome. Br J Cancer. 107:967–976. 2012. View Article : Google Scholar : PubMed/NCBI

16 

Cazes A, Lopez-Delisle L, Tsarovina K, Pierre-Eugene C, De Preter K, Peuchmaur M, Nicolas A, Provost C, Louis-Brennetot C, Daveau R, et al: Activated Alk triggers prolonged neurogenesis and Ret upregulation providing a therapeutic target in ALK-mutated neuroblastoma. Oncotarget. 5:2688–2702. 2014. View Article : Google Scholar : PubMed/NCBI

17 

Futami H and Sakai R: RET protein promotes non-adherent growth of NB-39-nu neuroblastoma cell line. Cancer Sci. 100:1034–1039. 2009. View Article : Google Scholar : PubMed/NCBI

18 

Meier R, Mühlethaler-Mottet A, Flahaut M, Coulon A, Fusco C, Louache F, Auderset K, Bourloud KB, Daudigeos E, Ruegg C, et al: The chemokine receptor CXCR4 strongly promotes neuroblastoma primary tumour and metastatic growth, but not invasion. PLoS One. 2:e10162007. View Article : Google Scholar : PubMed/NCBI

19 

Liberman J, Sartelet H, Flahaut M, Mühlethaler-Mottet A, Coulon A, Nyalendo C, Vassal G, Joseph JM and Gross N: Involvement of the CXCR7/CXCR4/CXCL12 axis in the malignant progression of human neuroblastoma. PLoS One. 7:e436652012. View Article : Google Scholar : PubMed/NCBI

20 

Wedge SR, Ogilvie DJ, Dukes M, Kendrew J, Chester R, Jackson JA, Boffey SJ, Valentine PJ, Curwen JO, Musgrove HL, et al: ZD6474 inhibits vascular endothelial growth factor signaling, angiogenesis, and tumor growth following oral administration. Cancer Res. 62:4645–4655. 2002.PubMed/NCBI

21 

Vidal M, Wells S, Ryan A and Cagan R: ZD6474 suppresses oncogenic RET isoforms in a Drosophila model for type 2 multiple endocrine neoplasia syndromes and papillary thyroid carcinoma. Cancer Res. 65:3538–3541. 2005. View Article : Google Scholar : PubMed/NCBI

22 

Wells SA Jr, Gosnell JE, Gagel RF, Moley J, Pfister D, Sosa JA, Skinner M, Krebs A, Vasselli J and Schlumberger M: Vandetanib for the treatment of patients with locally advanced or metastatic hereditary medullary thyroid cancer. J Clin Oncol. 28:767–772. 2010. View Article : Google Scholar : PubMed/NCBI

23 

Fox E, Widemann BC, Chuk MK, Marcus L, Aikin A, Whitcomb PO, Merino MJ, Lodish M, Dombi E, Steinberg SM, et al: Vandetanib in children and adolescents with multiple endocrine neoplasia type 2B associated medullary thyroid carcinoma. Clin Cancer Res. 19:4239–4248. 2013. View Article : Google Scholar : PubMed/NCBI

24 

Wells SA Jr, Robinson BG, Gagel RF, Dralle H, Fagin JA, Santoro M, Baudin E, Elisei R, Jarzab B, Vasselli JR, et al: Vandetanib in patients with locally advanced or metastatic medullary thyroid cancer: A randomized, double-blind phase III trial. J Clin Oncol. 30:134–141. 2012. View Article : Google Scholar : PubMed/NCBI

25 

Lee EQ, Kaley TJ, Duda DG, Schiff D, Lassman AB, Wong ET, Mikkelsen T, Purow BW, Muzikansky A, Ancukiewicz M, et al: A multicenter, phase II, randomized, noncomparative clinical trial of radiation and temozolomide with or without vandetanib in newly diagnosed glioblastoma patients. Clin Cancer Res. 21:3610–3618. 2015. View Article : Google Scholar : PubMed/NCBI

26 

Siegfried JM, Gubish CT, Rothstein ME, Henry C and Stabile LP: Combining the multitargeted tyrosine kinase inhibitor vandetanib with the antiestrogen fulvestrant enhances its antitumor effect in non-small cell lung cancer. J Thorac Oncol. 7:485–495. 2012. View Article : Google Scholar : PubMed/NCBI

27 

Gautschi O, Zander T, Keller FA, Strobel K, Hirschmann A, Aebi S and Diebold J: A patient with lung adenocarcinoma and RET fusion treated with vandetanib. J Thorac Oncol. 8:e43–e44. 2013. View Article : Google Scholar : PubMed/NCBI

28 

Ding X, Xiang L, Wang N, Zhao Z and Jin X, Sun Y, Duan W, Wang S and Jin X: Vandetanib-induced inhibition of neuroblastoma cell migration and invasion is associated with downregulation of the SDF-1/CXCR4 axis and matrix metalloproteinase 14. Oncol Rep. 31:1165–1174. 2014. View Article : Google Scholar : PubMed/NCBI

29 

Zage PE, Zeng L, Palla S, Fang W, Nilsson MB, Heymach JV and Zweidler-McKay PA: A novel therapeutic combination for neuroblastoma: The vascular endothelial growth factor receptor/epidermal growth factor receptor/rearranged during transfection inhibitor vandetanib with 13-cis-retinoic acid. Cancer. 116:2465–2475. 2010.PubMed/NCBI

30 

Sebaugh JL: Guidelines for accurate EC50/IC50 estimation. Pharm Stat. 10:128–134. 2011. View Article : Google Scholar : PubMed/NCBI

31 

Dai L, Cui X, Zhang X, Cheng L, Liu Y, Yang Y, Fan P, Wang Q, Lin Y, Zhang J, et al: SARI inhibits angiogenesis and tumour growth of human colon cancer through directly targeting ceruloplasmin. Nat Commun. 7:119962016. View Article : Google Scholar : PubMed/NCBI

32 

Dai L, Cheng L, Zhang X, Jiang Q, Zhang S, Wang S, Li Y, Chen X, Du T, Yang Y, et al: Plasmid-based STAT3-siRNA efficiently inhibits breast tumor growth and metastasis in mice. Neoplasma. 58:538–547. 2011. View Article : Google Scholar : PubMed/NCBI

33 

Yang C, Tan J, Zhu J, Wang S and Wei G: YAP promotes tumorigenesis and cisplatin resistance in neuroblastoma. Oncotarget. 8:37154–37163. 2017.PubMed/NCBI

34 

Michaelis M, Bliss J, Arnold SC, Hinsch N, Rothweiler F, Deubzer HE, Witt O, Langer K, Doerr HW, Wels WS, et al: Cisplatin-resistant neuroblastoma cells express enhanced levels of epidermal growth factor receptor (EGFR) and are sensitive to treatment with EGFR-specific toxins. Clin Cancer Res. 14:6531–6537. 2008. View Article : Google Scholar : PubMed/NCBI

35 

Rössler J, Odenthal E, Geoerger B, Gerstenmeyer A, Lagodny J, Niemeyer CM and Vassal G: EGFR inhibition using gefitinib is not active in neuroblastoma cell lines. Anticancer Res. 29:1327–1333. 2009.PubMed/NCBI

36 

Bakir S, Yazgan ÜC, Ibiloglu I, Elbey B, Kizil M and Kelle M: The protective effect of pomegranate extract against cisplatin toxicity in rat liver and kidney tissue. Arch Physiol Biochem. 121:152–156. 2015. View Article : Google Scholar : PubMed/NCBI

37 

Katanić J, Matić S, Pferschy-Wenzig EM, Kretschmer N, Boroja T, Mihailović V, Stanković V, Stanković N, Mladenović M, Stanić S, et al: Filipendula ulmaria extracts attenuate cisplatin-induced liver and kidney oxidative stress in rats: In vivo investigation and LC-MS analysis. Food Chem Toxicol. 99:86–102. 2017. View Article : Google Scholar : PubMed/NCBI

38 

Greco WR, Bravo G and Parsons JC: The search for synergy: A critical review from a response surface perspective. Pharmacol Rev. 47:331–385. 1995.PubMed/NCBI

39 

Minto CF, Schnider TW, Short TG, Gregg KM, Gentilini A and Shafer SL: Response surface model for anesthetic drug interactions. Anesthesiology. 92:1603–1616. 2000. View Article : Google Scholar : PubMed/NCBI

40 

Papangeli I, Kim J, Maier I, Park S, Lee A, Kang Y, Tanaka K, Khan OF, Ju H, Kojima Y, et al: MicroRNA 139-5p coordinates APLNR-CXCR4 crosstalk during vascular maturation. Nat Commun. 7:112682016. View Article : Google Scholar : PubMed/NCBI

41 

Liu H, Liu Y, Liu W, Zhang W and Xu J: EZH2-mediated loss of miR-622 determines CXCR4 activation in hepatocellular carcinoma. Nat Commun. 6:84942015. View Article : Google Scholar : PubMed/NCBI

42 

Geminder H, Sagi-Assif O, Goldberg L, Meshel T, Rechavi G, Witz IP and Ben-Baruch A: A possible role for CXCR4 and its ligand, the CXC chemokine stromal cell-derived factor-1, in the development of bone marrow metastases in neuroblastoma. J Immunol. 167:4747–4757. 2001. View Article : Google Scholar : PubMed/NCBI

43 

Airoldi I, Raffaghello L, Piovan E, Cocco C, Carlini B, Amadori A, Corrias MV and Pistoia V: CXCL12 does not attract CXCR4+ human metastatic neuroblastoma cells: Clinical implications. Clin Cancer Res. 12:77–82. 2006. View Article : Google Scholar : PubMed/NCBI

44 

Zhang L, Yeger H, Das B, Irwin MS and Baruchel S: Tissue microenvironment modulates CXCR4 expression and tumor metastasis in neuroblastoma. Neoplasia. 9:36–46. 2007. View Article : Google Scholar : PubMed/NCBI

45 

Catani MV, Corasaniti MT, Navarra M, Nisticò G, Finazzi-Agrò A and Melino G: gp120 induces cell death in human neuroblastoma cells through the CXCR4 and CCR5 chemokine receptors. J Neurochem. 74:2373–2379. 2000. View Article : Google Scholar : PubMed/NCBI

46 

Zhi Y, Duan Y, Zhou X, Yin X, Guan G, Zhang H, Dong Q and Yang K: NF-κB signaling pathway confers neuroblastoma cells migration and invasion ability via the regulation of CXCR4. Med Sci Monit. 20:2746–2752. 2014. View Article : Google Scholar : PubMed/NCBI

47 

Clift IC, Bamidele AO, Rodriguez-Ramirez C, Kremer KN and Hedin KE: β-Arrestin1 and distinct CXCR4 structures are required for stromal derived factor-1 to downregulate CXCR4 cell-surface levels in neuroblastoma. Mol Pharmacol. 85:542–552. 2014. View Article : Google Scholar : PubMed/NCBI

Related Articles

Journal Cover

April-2018
Volume 39 Issue 4

Print ISSN: 1021-335X
Online ISSN:1791-2431

Sign up for eToc alerts

Recommend to Library

Copy and paste a formatted citation
x
Spandidos Publications style
Li C, Yang C and Wei G: Vandetanib inhibits cisplatin‑resistant neuroblastoma tumor growth and invasion. Oncol Rep 39: 1757-1764, 2018
APA
Li, C., Yang, C., & Wei, G. (2018). Vandetanib inhibits cisplatin‑resistant neuroblastoma tumor growth and invasion. Oncology Reports, 39, 1757-1764. https://doi.org/10.3892/or.2018.6255
MLA
Li, C., Yang, C., Wei, G."Vandetanib inhibits cisplatin‑resistant neuroblastoma tumor growth and invasion". Oncology Reports 39.4 (2018): 1757-1764.
Chicago
Li, C., Yang, C., Wei, G."Vandetanib inhibits cisplatin‑resistant neuroblastoma tumor growth and invasion". Oncology Reports 39, no. 4 (2018): 1757-1764. https://doi.org/10.3892/or.2018.6255