Open Access

Co-expression of ING4 and P53 enhances hypopharyngeal cancer chemosensitivity to cisplatin in vivo

  • Authors:
    • Xin Ren
    • Hao Liu
    • Mingjie Zhang
    • Mengjun Wang
    • Shiyin Ma
  • View Affiliations

  • Published online on: July 27, 2016     https://doi.org/10.3892/mmr.2016.5552
  • Pages: 2431-2438
  • Copyright: © Ren et al. This is an open access article distributed under the terms of Creative Commons Attribution License.

Metrics: Total Views: 0 (Spandidos Publications: | PMC Statistics: )
Total PDF Downloads: 0 (Spandidos Publications: | PMC Statistics: )


Abstract

Hypopharyngeal cancer is a distinct type of malignant head and neck tumor, which exhibits low sensitivity to anti-cancer drugs. The importance of developing methods for reducing chemotherapy resistance, and improving and enhancing prognosis has previously been emphasized and is considered a challenge for effective clinical treatment of hypopharyngeal cancer. The current study investigated the effects of co‑expression of inhibitor of growth protein 4 (ING4) and P53, a tumor suppressor gene, on chemosensitivity to cisplatin in human hypopharyngeal cancer xenografts in vivo, and the potential molecular mechanisms involved. A tumor model was established by injecting athymic nude mice with FADU human hypopharyngeal cancer cells. Five days after intratumoral and peritumoral injections of an empty adenoviral vector (Ad), Ad‑ING4‑P53, cisplatin, or a combination of Ad‑ING4‑P53 and cisplatin (Ad‑ING4‑P53 + cisplatin) every other day for 5 days, the mice were euthanized and their tumors, livers, and kidneys were removed. The tumor weights were used to calculate the inhibition rate, and the expression levels of ING4 and P53 were detected by reverse transcription‑polymerase chain reaction. Additionally, apoptotic cells were detected using terminal deoxynucleotidyl transferase dUTP nick end labeling, and immunohistochemistry determined the levels ING4, P53, B‑cell lymphoma‑2 (Bcl‑2) and Bcl‑2 associated X protein (Bax) protein expression. The results demonstrated increased expression of ING4 and P53 in the Ad‑ING4‑P53 groups compared with PBS and Ad groups, indicating successful introduction of the genes into the tumor cells. Notably, the Ad‑ING4‑P53 + cisplatin group exhibited a higher inhibition rate compared with the four other groups. The results of immunohistochemistry analysis demonstrated that Bax expression was increased and Bcl‑2 was decreased in the Ad‑ING4‑P53 + cisplatin group. This suggested that the enhanced cisplatin chemosensitivity with Ad-ING4-P53 gene therapy in hypopharyngeal cancer xenografts may be associated with apoptosis induction through upregulation of Bax expression and downregulation of Bcl‑2. The results of the present study indicated that gene therapy combined with cisplatin treatment may be a promising treatment for human hypopharyngeal cancer.

Introduction

Head and neck squamous cell carcinoma (HNSCC) is one of the most common malignancies worldwide, and is characterized by high invasiveness, early metastasis, recurrence and difficult early detection (1,2). Hypopharyngeal cancer is a distinct type of malignant head and neck tumor currently treated by surgical procedures, radiotherapy and chemotherapy, which have numerous side effects, including loss of larynx function that severely affects quality of life. Despite the apparent advances in surgery in recent years, distant metastases and recurrence have remained concerns (3).

Tumor development is a multi-step process involving multiple genes, which includes the activation of proto-oncogenes and inactivation of tumor suppressor genes (4). With improved understanding of the molecular mechanisms underlying this process, gene therapy exhibits increasing potential for use as a novel cancer treatment. Hypopharyngeal cancer exhibits low sensitivity to anti-cancer drugs. Its strong resistance to various anti-tumor therapies and the unknown underlying mechanism lead to unfavorable prognosis and a low five-year survival rate for patients (5). Therefore, the importance of developing methods for avoiding chemotherapy resistance, and improving and enhancing prognosis has been emphasized, and is considered a challenge for effective clinical treatment of hypopharyngeal cancer.

Previous studies have demonstrated that gene therapy using a combination of two genes can promote tumor cell apoptosis. For example, Luo et al (6) reported that the adenovirus-mediated CD/TK double suicide gene, driven by a survivin promoter, specifically inhibits gastric cancer cells to a greater extent than that observed with a single suicide gene. Therefore, the effect of combined gene therapy on hypopharyngeal tumor survival was investigated.

The inhibitor of growth protein 4 (ING4) gene was originally identified by Shiseki et al (7) and later recognized as an important factor in tumor growth inhibition (8). It is expressed in all cells, including normal tissues. Extensive studies have demonstrated that ING4 is critical for gene transcription, cell proliferation, apoptosis and senescence, cell contact inhibition, DNA damage repair, and tumor invasion and metastasis (912).

Recent studies have packaged the ING4 gene into an adenoviral vector (Ad-ING4) for introduction into various human tumor cells, including malignant melanoma (13), breast cancer (14), human lung adenocarcinoma (15) and osteosarcoma (16). These studies demonstrated significantly higher growth inhibition and apoptosis in tumor cells with Ad-ING4 compared with cells infected with the empty vector, indicating that ING4 inhibits the growth of tumor cells and induces their apoptosis. A previous study demonstrated that there were decreased expression levels of ING4 in HNSCC and concluded that it is important in cancer cell apoptosis and is anti-proliferative (17). However, its function and mechanism of action remain unknown and require further study.

P53, the first member of the P53 family to be identified, is associated with various types of cancer, including sporadic cancers, which are correlated with mutations in somatic cells (18). Although wild-type P53 inhibits tumor development and progression, a previous study demonstrated that P53 is generally mutated in tumor cells to induce and promote tumor development (19). Gene therapy with wild-type P53 and other P53 family members was observed to be effective and safe for the treatment of pulmonary metastatic tumors from hepatocellular carcinoma (20) and inhibited proliferation and apoptosis in osteosarcoma cell lines (21). Furthermore, previous investigation demonstrated that ING4 induces apoptosis through a P53-dependent pathway (22,23).

The present study used combination gene therapy with ING4 and P53 to treat hypopharyngeal cancer in vivo. Mutations identified in 40–70% of HNSCC tumors occur in P53 (24), providing a theoretical basis for adenovirus-mediated combination gene therapy to inhibit hypopharyngeal cancer cell growth and proliferation. Thus, it was hypothesized that combination treatment with ING4 and P53 tumor suppressors would enhance tumor chemosensitivity.

Materials and methods

Cell lines

The FADU human hypopharyngeal cancer cell line was purchased from American Type Culture Collection (Manassas, VA, USA) and cultured in Dulbecco's modified Eagle's medium (DMEM; Gibco; Thermo Fisher Scientific, Inc., Waltham, MA, USA) with 10% fetal bovine serum (Zhejiang Tianhang Biotechnology Co., Ltd. (Huzhou, China) and 1% antibiotic solution (100X penicillin and streptomycin; Beyotime Institute of Biotechnology, Haimen, China) at 37°C in a humidified atmosphere composed of 95% air and 5% CO2.

Virus propagation and purification

Adenoviral vectors with the ING4 and P53 genes were provided by Professor Jicheng Yang (Soochow University, Suzhou, China). QBI 293A cells (American Type Culture Collection) with the adenoviral vectors to amplify the recombinant virus promoter, and specific steps and titers were measured by conventional methods as described previously (16). The QBI 293A cells were cultured as described for the FADU cells, however, in RPMI 1640 (Gibco; Thermo Fisher Scientific, Inc.) rather than DMEM.

Animals

Specific-pathogen free BALB/c nu/nu nude mice (25 males; weight, 22–25 g; age, 5–6 weeks) were obtained from the Changzhou Cavens Laboratory Animal Co. [Changzhou, China; certificate no. SCXK (Su) 2011-0003 Su regulatory certificate no. 201403734]. They were maintained and used for the experiments performed at the Laboratory Animal Center of Bengbu Medical University (Anhui, China; certificate no. Wan SYXK 2012-002). The current study was approved by the appropriate ethical review boards for the use of laboratory animals. The animals were fed as standard with access to drinking water. The temperature was maintained at 23–28°C and relative humidity was 40–60%, with a natural light/dark cycle.

Establishment of tumor models

Each mouse was injected subcutaneously with 1×106 human FADU hypopharyngeal cancer cells in the axilla of the right anterior limb. The tumor dimensions were measured 2–3 times per week with a caliper, and the tumor volume was calculated as follows: Tumor size = a×b2/2, where a and b represent the larger and smaller of the two dimensions, respectively.

Experimental design and preparation of cisplatin

The mice were randomly assigned to five groups (five mice per group) when the tumors developed to a mean volume of 60–80 mm3 after ~20 days. The xenograft tumor-bearing mice were intra-tumorally and peritumorally injected with phosphate-buffered saline (PBS control), empty adenoviral vector [Ad; 0.1 ml, 1×108 plaque forming units (pfu)], Ad-ING4-P53 (0.1 ml, 1×108 pfu), cisplatin (0.1 ml, 300 µg), or Ad-ING4-P53 (0.1 ml, 1×108 pfu) and cisplatin (0.1 ml, 300 µg) every other day for five days. Cisplatin (3 mg; Qilu Pharmaceutical Co., Ltd., Jinan, China) was dissolved in 1 ml normal saline and used at 3 µg/µl in all experiments.

Five days after treatment, the mice were sacrificed by cervical dislocation. The transplanted tumors were removed and weighed to calculate the inhibition rate as follows: Inhibition rate = (1-mean experimental tumor weight/mean control tumor weight)×100%. Based on the inhibition rate, the combined effect was evaluated from the Q values (25) as follows: Q value = E (A + B)/(EA + EB − EA × EB), where EA and EB are the effects of molecules A and B alone, respectively, E (A + B) is the combined effect, and the denominator represents the expected effects when the two are combined. Molecules A and B are interpreted as having additive effects when Q=1±0.15, synergistic effects when Q>1.15, and antagonistic effects when Q<0.85.

Reverse transcription-polymerase chain reaction (RT-PCR)

Gene transcript levels were detected by RT-PCR analysis. Total RNA was extracted from the xenografted tumors using TRIzol according to the manufacturer's instructions (Invitrogen; Thermo Fisher Scientific, Inc.). The RT-PCR reactions were performed using a RevertAid First Strand cDNA Synthesis kit (Thermo Fisher Scientific, Inc.) to generate cDNA and then a PCR Master mix (2X; Thermo Fisher Scientific, Inc.) in an ABI StepOne™ Real-Time PCR System (Applied Biosystems; Thermo Fisher Scientific, Inc.) by using degenerate primers (Sangon Biotech Co., Ltd., Shanghai, China) (Table I). The thermocycling conditions were as follows: 95°C for 3 min; 40 cycles of 95°C for 30 sec, 61.5°C, 52.5°C, 59°C or 58°C for 30 sec (for ING4, P53, Bax and Bcl-2, respectively) and 72°C for 1 min; followed by 72°C for 10 min. GAPDH served as the control. The PCR products were separated on 1.5% agarose gel.

Table I

Reverse transcription-polymerase chain reaction primers.

Table I

Reverse transcription-polymerase chain reaction primers.

NamePrimer sequenceSize (bp)
GADPH240
 F 5′-TGATGACATCAAGAAGGTGGTGAA-3′
 R 5′-TCCTTGGAGGCCATGTGGGCC-3′
ING4750
 F 5′-TAGAGATCTACCATGGCTGCTGGGATGTATTTGG-3′
 R 5′-ACCGTCGACCCTATTTCTTCTTCCGTTCTTG-3′
P53259
 F 5′-CCTCCTCAGCATCTTATCCG-3′
 R 5′-CACAAACACGCACCTCAAA-3′
Bcl-2304
 F 5′-TTCTTTGAGTTCGGTGGGGTC-3′
 R 5′-TGCATATTTGTTTGGGGCAGG-3′
Bax257
 F 5′-TCCACCAAGAAGCTGAGCGAG-3′
 R 5′-GTCCAGCCCATGATGGTTCT-3′

[i] ING4, inhibitor of growth protein 4; Bcl-2, B-cell lymphoma-2; Bax, Bcl-2 associated X protein.

Hematoxylin and eosin (H&E) staining

The mice were sacrificed by cervical dislocation 5 days after drug administration, and the xenografted tumors, livers and kidneys were removed to observe the distribution of tumor cells, and the tumor metastasis and cytotoxicity of the livers and kidneys. Tissue sections were incubated overnight in neutral formalin buffer (10%) and then stored in ethanol and embedded in paraffin. Cross-sections (4 µm) were stained with H&E and observed under a microscope (BX43; Olympus Corporation, Tokyo, Japan), and tumor cells were identified by the following characteristics: Small volume, condensed cytoplasm, small nuclei, condensed and fractured chromatin, nuclei migrated to the cell edge, or apoptotic bodies with karyorrhexis.

Terminal deoxynucleotidyl transferase dUTP nick end labeling (TUNEL) assay

Sections of tumor tissue were fixed in 10% neutral formalin buffer according to the standard procedure and then stained using an In Situ Cell Apoptosis Detection Kit IV following the manufacturer's instructions (Sigma-Aldrich, St. Louis, MO, USA) using conventional methods as described previously (26). The sections were analyzed under a confocal microscope (×200). Yellow granules in the nucleus indicated apoptotic cells.

Immunohistochemistry in xenografted tumors

The expression of ING4, P53, B-cell lymphoma-2 (Bcl-2), and Bcl-2 associated X protein (Bax) in the xenografted tumors was analyzed by immunohistochemistry using an Ultrasensitive™ SP kit (Fuzhou Maixin Biotech Co., Ltd., Fuzhou, China). Tissue sections were deparaffinized in dimethyl benzene and dehydrated by an alcohol gradient. The sections were incubated in 3% H2O2 for 10 min to block inactivated endogenous peroxidase. For antigen retrieval, the sections were placed in 0.01 M citrate buffer (pH 6.0) and boiled at 95°C for 15–20 min, cooled at room temperature for 20 min, and washed with cold water to facilitate cooling. Then sections were sealed in normal bovine serum (Zhejiang Tianhang Biotechnology Co., Ltd.) for 10 min at 37°C, and then the bovine serum was removed. The polyclonal rabbit anti-mouse antibodies were incubated for 1 h at room temperature and were as follows: Anti-ING4 (1:50; cat. no. 16188-1-AP), anti-P53 (1:100; cat. no. 10442-1-AP), anti-Bcl-2 (1:50; cat. no. 12789-1-AP) and Bax (1:50; cat. no. 23931-1-AP; all from Proteintech Group, Inc., Chicago, IL, USA) Biotinylated goat anti-rabbit secondary antibodies (1:40; Proteintech Group, Inc.; cat. no. SA00001-2) were then incubated for 20 min at room temperature. Sections were subsequently incubated in peroxidase substrate mixing liquid (Fuzhou Maixin Biotech Co., Ltd.) and washed in deionized water. Finally, the sections were counterstained with hematoxylin, dehydrated with deionized water, dried and sealed. They were observed by microscopy (BX43). Positive gene expression was indicated by the presence of yellow diaminobenzidine precipitates.

Statistical analysis

Data were analyzed by SPSS for Windows (version 17.0; SPSS, Inc., Chicago, IL, USA) and are presented as the mean ± standard error of the mean. Differences between groups were evaluated by one-way analysis of variance (ANOVA), two-way ANOVA, and Dunnett's multiple comparisons test. P<0.05 was considered statistically significant.

Results

Establishment of tumor models

Subcutaneous solid nodules developed gradually for 6–7 days post-injection. At ~20 days later, the mean tumor volume was 60–80 mm3. All 25 nude mice were tumorigenic, with a tumor formation rate of 100%. During treatment, the mice exhibited dry skin, however there were no significant changes in their stool color, appetite or behavior. The mice treated with cisplatin and a combination of Ad-ING4-P53 and cisplatin (Ad-ING4-P53 + cisplatin) exhibited weight loss, which was most notable in the cisplatin group. Following sacrifice of the mice, no significant macroscopic or microscopic lesions were observed in the organs.

Effect of Ad-ING4-P53 + cisplatin on FADU cell inhibition

Following establishment of the tumor models, the xenograft tumor-bearing mice were intratumorally and peritumorally injected with PBS, Ad, Ad-ING4-P53, cisplatin, or Ad-ING4-P53 + cisplatin every other day for five days. We measured the xenograft tumor volumes and weighed the mice every other day (Fig. 1A and B) and removed and weighed the tumors five days after treatment (Fig. 1C).

Compared with the cisplatin and Ad-ING4-P53 groups, Ad-ING4-P53 + cisplatin significantly inhibited the growth of FADU hypopharyngeal cancer cells in nude mice bearing transplantation tumors (P<0.05; Fig. 1D) and exerted a synergistic effect (Q=1.19).

Expression of ING4 and P53

Results of the RT-PCR analysis of ING4 and P53 gene expression levels are demonstrated in Fig. 2. GADPH was used as a reference gene. The Ad-ING4-P53 and Ad-ING4-P53 + cisplatin groups demonstrated strong positive ING4/P53 bands compared with the PBS, Ad and cisplatin groups. Furthermore, immunohistochemical analysis demonstrated no obvious expression of ING4/P53 in the PBS, Ad and cisplatin groups, whereas the Ad-ING4-P53 and Ad-ING4-P53 + cisplatin groups demonstrated positive ING4 and P53 expression (Fig. 3).

Apoptotic gene expression changes in xenografted tumors

To determine the potential molecular mechanisms of apoptosis induction, Bcl-2 and Bax expression levels were detected in the tumor tissue using RT-PCR. Compared with the PBS group, the gene expression of the Ad group changed only marginally, whereas the Ad-ING4-P53 and Ad-ING4-P53 + cisplatin groups demonstrated strongly positive Bax expression and negative Bcl-2 expression, with a greater change in the Ad-ING4-P53 + cisplatin group (Fig. 2) compared with the PBS and Ad groups. These results were further supported by immunohistochemical analysis which demonstrated the Ad-ING4-P53 + cisplatin group exhibited increased ING4, P53 and Bax expression levels and decreased Bcl-2 expression compared to other groups (Fig. 3).

Enhanced tumor apoptosis by Ad-ING4-P53 + cisplatin

Sections of xenografted hypopharyngeal tumors from nude mice were stained by H&E (Fig. 4A), and the tumor cells exhibited a nest-like distribution and disordered arrangement. In the PBS and Ad groups, the tumor cells were closely arranged with complete and atypical structures. By contrast, the Ad-ING4-P53, cisplatin and Ad-ING4-P53 + cisplatin groups exhibited a greater degree of tumor cell apoptosis, characterized by incomplete cell membranes, condensed cytoplasm, pyknotic or cracking nuclei, and cavity-shaped organization. The Ad-ING4-P53 group and cisplatin group demonstrated similar characteristic, whereas the apoptotic effect was more obvious in the Ad-ING4-P53 + cisplatin group.

TUNEL analysis demonstrated the apoptotic cells as small, with condensed nuclei, circumscribed nuclear membranes and yellow granules in the nuclei (Fig. 4A). Comparison of the five treatment groups indicated that Ad-ING4-P53, cisplatin and Ad-ING4-P53 + cisplatin accelerated apoptosis relative to PBS and Ad, with the greatest effect exerted by treatment with Ad-ING4-P53 + cisplatin.

Toxicity of gene therapy and cisplatin

H&E-stained liver and kidney sections from human hypopharyngeal tumor-bearing nude mice were observed microscopically (Fig. 4B). The PBS and cisplatin groups exhibited normal liver tissue, with clear lobule structures and orderly hepatic cords. However, the Ad, Ad-ING4-P53, and Ad-ING4-P53 + cisplatin groups exhibited irregularly bleeding necrotic areas, and their liver cell structures did not demonstrate marked inflammatory cell infiltration. However, the kidney microstructures were in good condition for all groups, with no pathological changes, such as blood extravasation or cell necrosis. Thus, the adenoviral vector induced liver toxicity.

Discussion

Cisplatin is the most commonly used chemotherapeutic drug, particularly for the treatment of head and neck cancer. However, the development of cisplatin resistance has limited its widespread clinical use (2729). Therefore, identification of high-efficiency chemosensitization drugs to improve the efficiency of chemotherapy is an important direction of current research.

The ING4 gene was identified as an important tumor growth inhibition factor (8). Further studies demonstrated that ING4 exerts specific anti-tumor effects through various pathways and can induce tumor cell apoptosis. Zhang et al (30) established several HepG-2 hepatocellular carcinoma cell lines stably expressing ING4 and observed that ING4 inhibited HepG-2 cell growth and improved the sensitivity of liver cancer cells to DNA damage reagents, including adriamycin. Another important factor, P53, is regarded as an important factor in tumor activation and an essential gene for genome integrity (31). Kraljević Pavelić et al (32) introduced the P53 gene into Hep-2 and CAL27 HNSCC cell lines and demonstrated that p53 overexpression at sub-cytotoxic levels enhanced the activity of low doses of cisplatin and methotrexate through changes in the cell cycle.

Resistance to multiple anti-tumor therapies arises through complex mechanisms, and tumor cell apoptosis resistance is generally understood to be a major mechanism of multi-drug resistance. The majority of chemotherapy drugs act by inducing tumor cell apoptosis. The Bcl-2 gene inhibits apoptosis caused by various factors, including carcinogens and radioactive rays, abnormally extends cell survival time, promotes the accumulation of mutations and increases resistance to immune system monitoring (33). A previous study demonstrated an association between excessive expression of Bcl-2, and expression of tumor cell drug resistance genes and inhibition of apoptosis, which leads to drug resistance (34). Bax is an apoptosis-promoting gene in the Bcl-2 family. In vivo, Bax/Bax cognate dimer formation promotes cell apoptosis, whereas Bcl-2/Bax heterologous dimers inhibit apoptosis (35).

The present study demonstrated that the Ad, Ad-ING4-P53, and Ad-ING4-P53 + cisplatin groups exhibited liver toxicity, however they exerted no effect on kidney cells, consistent with previous demonstrations that adenoviral vectors damage liver function (36,37). Haisma et al (38) demonstrated that liver toxicity arises from removal of Kupffer cells by adenoviral vectors, potentially as an immune response to the virus and its transduction gene products (39). Future studies must investigate whether changes in the adenovirus vector structure or simultaneous of administration liver-protection drugs would abrogate the observed liver toxicity.

The current study was designed to investigate the chemo-sensitivity of a recombinant adenovirus co-expressing ING4 and P53, and to analyze the potential mechanisms underlying this process. The results of the present study demonstrated that Ad-ING4-P53 + cisplatin significantly inhibited the growth of FADU hypopharyngeal cancer cells in nude mice bearing transplanted tumors compared with the effect of cisplatin or Ad-ING4-P53 alone (P<0.05) and a synergistic effect between Ad-ING4-P53 and cisplatin was observed (Q=1.19). Immunohistochemical analysis of the expression of associated factors in transplanted tumors further indicated that Ad-ING4-P53 + cisplatin strongly increased Bax expression and decreased Bcl-2 expression compared with the levels observed with cisplatin or Ad-ING4-P53 alone, which is consistent with previous findings by Zhu et al (40). Thus, the combination therapy may increase chemosensitivity through the Bcl-2/Bax pathway; however, elucidation of the specific underlying mechanism requires further research.

In conclusion, the results of the current study demonstrated that Ad-ING4-P53 improves the chemosensitivity of transplanted human hypopharyngeal tumors, potentially by increasing and decreasing the expression levels of Bax and Bcl-2, respectively, to induce tumor cell apoptosis. The co-expression of ING4 and P53 genes via a recombinant adenovirus synergistically enhanced the anti-tumor effects with simultaneous chemotherapy for human hypopharyngeal cancer in vivo. Further investigation is required to elucidate the specific underlying mechanism of action in vitro and in vivo, however, this work provides an experimental basis for these future studies and for clinical applications.

Acknowledgments

This study was supported by the Natural Science Research Project of Colleges and Universities of Anhui Province (grant no. KJ2013A193).

Abbreviations:

Ad

adenoviral vector

ING4

inhibitor of growth protein 4

References

1 

Genden EM, Ferlito A, Bradley PJ, Rinaldo A and Scully C: Neck disease and distant metastases. Oral Oncol. 39:207–212. 2003. View Article : Google Scholar : PubMed/NCBI

2 

Torre LA, Bray F, Siegel RL, Ferlay J, Lortet-Tieulent J and Jemal A: Global cancer statistics, 2012. CA Cancer J Clin. 65:87–108. 2015. View Article : Google Scholar : PubMed/NCBI

3 

Keereweer S, Kerrebijn JD, Al-Mamgani A, Sewnaik A, Baatenburg de Jong RJ and van Meerten E: Chemoradiation for advanced hypopharyngeal carcinoma: A retrospective study on efficacy, morbidity and quality of life. Eur Arch Otorhinolaryngol. 269:939–946. 2012. View Article : Google Scholar :

4 

Hanna NN, Mauceri HJ, Wayne JD, Hallahan DE, Kufe DW and Weichselbaum RR: Virally directed cytosine deaminase/5-fluo-rocytosine gene therapy enhances radiation response in human cancer xenografts. Cancer Res. 57:4205–4209. 1997.PubMed/NCBI

5 

Thurfjell N, Coates PJ, Boldrup L, Lindgren B, Bäcklund B, Uusitalo T, Mahani D, Dabelsteen E, Dahlqvist A, Sjöström B, et al: Function and importance of p63 in normal oral mucosa and squamous cell carcinoma of the head and neck. Adv Otorhinolaryngol. 62:49–57. 2005.

6 

Luo XR, Li JS, Niu Y and Miao L: Adenovirus-mediated double suicide gene selectively kills gastric cancer cells. Asian Pac J Cancer Prev. 13:781–784. 2012. View Article : Google Scholar : PubMed/NCBI

7 

Shiseki M, Nagashima M, Pedeux RM, Kitahama-Shiseki M, Miura K, Okamura S, Onogi H, Higashimoto Y, Appella E, Yokota J and Harris CC: p29ING4 and p28ING5 bind to p53 and p300, and enhance p53 activity. Cancer Res. 63:2373–2378. 2003.PubMed/NCBI

8 

Garkavtsev I, Kozin SV, Chernova O, Xu L, Winkler F, Brown E, Barnett GH and Jain RK: The candidate tumour suppressor protein ING4 regulates brain tumour growth and angiogenesis. Nature. 428:328–332. 2004. View Article : Google Scholar : PubMed/NCBI

9 

Unoki M, Shen JC, Zheng ZM and Harris CC: Novel splice variants of ING4 and their possible roles in the regulation of cell growth and motility. J Biol Chem. 281:34677–34686. 2006. View Article : Google Scholar : PubMed/NCBI

10 

Colla S, Tagliaferri S, Morandi F, Lunghi P, Donofrio G, Martorana D, Mancini C, Lazzaretti M, Mazzera L, Ravanetti L, et al: The new tumor-suppressor gene inhibitor of growth family member 4 (ING4) regulates the production of proangiogenic molecules by myeloma cells and suppresses hypoxia-inducible factor-1 alpha (HIF-1alpha) activity: Involvement in myeloma-induced angio-genesis. Blood. 110:4464–4475. 2007. View Article : Google Scholar : PubMed/NCBI

11 

Shen JC, Unoki M, Ythier D, Duperray A, Varticovski L, Kumamoto K, Pedeux R and Harris CC: Inhibitor of growth 4 suppresses cell spreading and cell migration by interacting with a novel binding partner, liprin alpha1. Cancer Res. 67:2552–2558. 2007. View Article : Google Scholar : PubMed/NCBI

12 

Li J, Martinka M and Li G: Role of ING4 in human melanoma cell migration, invasion and patient survival. Carcinogenesis. 29:1373–1379. 2008. View Article : Google Scholar : PubMed/NCBI

13 

Cai L, Li X, Zheng S, Wang Y, Wang Y, Li H, Yang J and Sun J: Inhibitor of growth 4 is involved in melanomagenesis and induces growth suppression and apoptosis in melanoma cell line M14. Melanoma Res. 19:1–7. 2009. View Article : Google Scholar : PubMed/NCBI

14 

Wei Q, He W, Lu Y, Yao J and Cao X: Effect of the tumor suppressor gene ING4 on the proliferation of MCF-7 human breast cancer cells. Oncol Lett. 4:438–442. 2012.PubMed/NCBI

15 

Huang J, Yang J, Ling C, Zhao D, Xie Y and You Z: The mechanism of inhibition effect of adenovirus-mediated ING4 on human lung adenocarcinoma xenografts in nude mice. Zhongguo Fei Ai Za Zhi. 17:142–147. 2014.In Chinese. PubMed/NCBI

16 

Xu M, Xie Y, Sheng W, Miao J and Yang J: Adenovirus-mediated ING4 gene transfer in osteosarcoma suppresses tumor growth via induction of apoptosis and inhibition of tumor angiogenesis. Technol Cancer Res Treat. 14:369–378. 2015. View Article : Google Scholar

17 

Gunduz M, Nagatsuka H, Demircan K, Gunduz E, Cengiz B, Ouchida M, Tsujigiwa H, Yamachika E, Fukushima K, Beder L, et al: Frequent deletion and down-regulation of ING4, a candidate tumor suppressor gene at 12p13, in head and neck squamous cell carcinomas. Gene. 356:109–117. 2005. View Article : Google Scholar : PubMed/NCBI

18 

Vijayaraman KP, Veluchamy M, Murugesan P, Shanmugiah KP and Kasi PD: p53 exon 4 (codon 72) polymorphism and exon 7 (codon 249) mutation in breast cancer patients in southern region (Madurai) of Tamil Nadu. Asian Pac J Cancer Prev. 13:511–516. 2012. View Article : Google Scholar : PubMed/NCBI

19 

Levine AJ, Finlay CA and Hinds PW: P53 is a tumor suppressor gene. Cell. 116(Suppl 2): S67–S69, 1 p following S69. 2004. View Article : Google Scholar : PubMed/NCBI

20 

Yu M, Chen W and Zhang J: p53 gene therapy for pulmonary metastasis tumor from hepatocellular carcinoma. Anticancer Drugs. 21:882–884. 2010. View Article : Google Scholar : PubMed/NCBI

21 

Oshima Y, Sasaki Y, Negishi H, Idogawa M, Toyota M, Yamashita T, Wada T, Nagoya S, Kawaguchi S, Yamashita T and Tokino T: Antitumor effect of adenovirus-mediated p53 family gene transfer on osteosarcoma cell lines. Cancer Biol Ther. 6:1058–1066. 2007. View Article : Google Scholar : PubMed/NCBI

22 

Russell M, Berardi P, Gong W and Riabowol K: Grow-ING, Age-ING and Die-ING: ING proteins link cancer, senescence and apoptosis. Exp Cell Res. 312:951–961. 2006. View Article : Google Scholar : PubMed/NCBI

23 

Soliman MA and Riabowol K: After a decade of study-ING, a PHD for a versatile family of proteins. Trends Biochem Sci. 32:509–519. 2007. View Article : Google Scholar : PubMed/NCBI

24 

Sozzi G, Miozzo M, Donghi R, Pilotti S, Cariani CT, Pastorino U, Della Porta G and Pierotti MA: Deletions of 17p and p53 mutations in preneoplastic lesions of the lung. Cancer Res. 52:6079–6082. 1992.PubMed/NCBI

25 

Jin ZJ: Addition in drug combination (author's transl). Zhongguo Yao Li Xue Bao. 1:70–76. 1980.In Chinese. PubMed/NCBI

26 

Yin W, Zhang J, Jiang Y and Juan S: Combination therapy with low molecular weight heparin and Adriamycin results in decreased breast cancer cell metastasis in CH mice. Exp Ther Med. 8:1213–1218. 2014.PubMed/NCBI

27 

Chan DA, Sutphin PD, Nguyen P, Turcotte S, Lai EW, Banh A, Reynolds GE, Chi JT, Wu J, Solow-Cordero DE, et al: Targeting GLUT1 and the Warburg effect in renal cell carcinoma by chemical synthetic lethality. Sci Transl Med. 3:94ra702011. View Article : Google Scholar : PubMed/NCBI

28 

Shimanishi M, Ogi K, Sogabe Y, Kaneko T, Dehari H, Miyazaki A and Hiratsuka H: Silencing of GLUT-1 inhibits sensitization of oral cancer cells to cisplatin during hypoxia. J Oral Pathol Med. 42:382–388. 2013. View Article : Google Scholar

29 

Wang YD, Li SJ and Liao JX: Inhibition of glucose transporter 1 (GLUT1) chemosensitized head and neck cancer cells to cisplatin. Technol Cancer Res Treat. 12:525–535. 2013.PubMed/NCBI

30 

Zhang X, Xu LS, Wang ZQ, Wang KS, Li N, Cheng ZH, Huang SZ, Wei DZ and Han ZG: ING4 induces G2/M cell cycle arrest and enhances the chemosensitivity to DNA-damage agents in HepG2 cells. FEBS Lett. 570:7–12. 2004. View Article : Google Scholar : PubMed/NCBI

31 

Zhou X, Gu Y and Zhang SL: Association between p53 codon 72 polymorphism and cervical cancer risk among Asians: A HuGE review and meta-analysis. Asian Pac J Cancer Prev. 13:4909–4914. 2012. View Article : Google Scholar : PubMed/NCBI

32 

Kraljević Pavelić S, Marjanović M, Poznić M and Kralj M: Adeno-virally mediated p53 overexpression diversely influence the cell cycle of HEp-2 and CAL 27 cell lines upon cisplatin and methotrexate treatment. J Cancer Res Clin Oncol. 135:1747–1761. 2009. View Article : Google Scholar

33 

Rudin CM and Thompson CB: Apoptosis and disease: Regulation and clinical relevance of programmed cell death. Ann Rev Med. 48:267–281. 1997. View Article : Google Scholar : PubMed/NCBI

34 

Tse C, Shoemaker AR, Adickes J, Anderson MG, Chen J, Jin S, Johnson EF, Marsh KC, Mitten MJ, Nimmer P, et al: ABT-263: A potent and orally bioavailable Bcl-2 family inhibitor. Cancer Res. 68:3421–3428. 2008. View Article : Google Scholar : PubMed/NCBI

35 

Cosulich SC, Savory PJ and Clarke PR: Bcl-2 regulates amplification of caspase activation by cytochrome c. Curr Biol. 9:147–150. 1999. View Article : Google Scholar : PubMed/NCBI

36 

Lozier JN, Csako G, Mondoro TH, Krizek DM, Metzger ME, Costello R, Vostal JG, Rick ME, Donahue RE and Morgan RA: Toxicity of a first-generation adenoviral vector in rhesus macaques. Hum Gene Ther. 13:113–124. 2002. View Article : Google Scholar : PubMed/NCBI

37 

Li Y, Shao JY, Liu RY, Zhou L, Chai LP, Li HL, Han HY, Huang BJ, Zeng MS, Zhu XF, et al: Evaluation of long-term toxicity of Ad/hIFN-, an Adenoviral vector encoding the human interferon-gamma gene, in nonhuman primates. Hum Gene Ther. 19:827–839. 2008. View Article : Google Scholar : PubMed/NCBI

38 

Haisma HJ, Boesjes M, Beerens AM, van der Strate BW, Curiel DT, Plüddemann A, Gordon S and Bellu AR: Scavenger receptor A: A new route for adenovirus 5. Mol Pharm. 6:366–374. 2009. View Article : Google Scholar : PubMed/NCBI

39 

Lieber A, He CY, Meuse L, Schowalter D, Kirillova I, Winther B and Kay MA: The role of Kupffer cell activation and viral gene expression in early liver toxicity after infusion of recombinant adenovirus vectors. J Virol. 71:8798–8807. 1997.PubMed/NCBI

40 

Zhu Y, Lv H, Xie Y, Sheng W, Xiang J and Yang J: Enhanced tumor suppression by an ING4/IL-24 bicistronic adenovirus-mediated gene cotransfer in human non-small cell lung cancer cells. Cancer Gene Ther. 18:627–636. 2011. View Article : Google Scholar : PubMed/NCBI

Related Articles

Journal Cover

September-2016
Volume 14 Issue 3

Print ISSN: 1791-2997
Online ISSN:1791-3004

Sign up for eToc alerts

Recommend to Library

Copy and paste a formatted citation
x
Spandidos Publications style
Ren X, Liu H, Zhang M, Wang M and Ma S: Co-expression of ING4 and P53 enhances hypopharyngeal cancer chemosensitivity to cisplatin in vivo. Mol Med Rep 14: 2431-2438, 2016
APA
Ren, X., Liu, H., Zhang, M., Wang, M., & Ma, S. (2016). Co-expression of ING4 and P53 enhances hypopharyngeal cancer chemosensitivity to cisplatin in vivo. Molecular Medicine Reports, 14, 2431-2438. https://doi.org/10.3892/mmr.2016.5552
MLA
Ren, X., Liu, H., Zhang, M., Wang, M., Ma, S."Co-expression of ING4 and P53 enhances hypopharyngeal cancer chemosensitivity to cisplatin in vivo". Molecular Medicine Reports 14.3 (2016): 2431-2438.
Chicago
Ren, X., Liu, H., Zhang, M., Wang, M., Ma, S."Co-expression of ING4 and P53 enhances hypopharyngeal cancer chemosensitivity to cisplatin in vivo". Molecular Medicine Reports 14, no. 3 (2016): 2431-2438. https://doi.org/10.3892/mmr.2016.5552