Open Access

Combined effect of recombinant human adenovirus p53 and curcumin in the treatment of liver cancer

  • Authors:
    • Juan Qu
    • Wei Lu
    • Ming Chen
    • Wei Gao
    • Cong Zhang
    • Bin Guo
    • Jizhi Yang
  • View Affiliations

  • Published online on: August 26, 2020     https://doi.org/10.3892/etm.2020.9145
  • Article Number: 18
  • Copyright: © Qu 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

The development of an effective therapeutic intervention for liver cancer is a worldwide challenge that remains to be adequately addressed. Of note, TP53, which encodes the p53 protein, is an important tumor suppressor gene, 61% of TP53 is functionally inactivated in liver cancer. Recombinant human adenovirus p53 (rAd‑p53) is the first commercial product that has been used for gene therapy. In the present study, the combined mechanistic effects of rAd‑p53 and curcumin, a naturally occurring compound with previously reported anti‑inflammatory, antioxidant and anti‑cancer properties, were assessed in liver cancer cells, using HepG2 cells as the model cell line. The administration of either curcumin or rAd‑p53 promoted apoptosis, suppressed epithelial‑mesenchymal transition (EMT) and blocked G2/M phase progression in HepG2 cells, which were potentiated further when both agents were applied together. Combined rAd‑p53 and curcumin treatment resulted in higher p53 (P<0.01) and p21 (P<0.01) expression compared with rAd‑p53 or curcumin were added alone, suggesting an additive effect on TP53 expression. Additionally, curcumin and rAd‑p53 were demonstrated to regulate the activation of mitogen‑activated protein kinases (MAPKs) ERK1/2, p38 MAPK and JNK. These results indicated that the combination of rAd‑p53 with curcumin synergistically potentiates apoptosis and inhibit EMT compared with either rAd‑p53 or curcumin treatment alone via the regulation of TP53 regulation. Mechanistically, this effect on TP53 expression may involve the ERK1/2, p38 MAPK and JNK signaling pathways. The current study provides new insights that can potentially advance the development of therapeutic strategies for liver cancer treatment.

Introduction

Liver cancer currently ranks as the third most common cause of mortality associated cancer worldwide, with >600,000 deaths reported annually (1,2). Liver cancer commonly occurs in patients with a history of chronic liver conditions, including hepatitis B and C viral infections, alcoholic or non-alcoholic liver disease, fatty liver disease and chronic liver disease that is caused by aflatoxin poisoning (3), in a vicious cycle of liver injury, regeneration and inflammation (1). Since effective clinical diagnosis and treatment of liver cancer is typically hindered by high rates of recurrence and metastasis (2,3), it is of importance to develop innovative therapeutic strategies for the diagnosis and treatment of liver cancer.

TP53 is an important human tumor suppressor gene that is present on chromosome 17p13.1(4), which contains 11 exons that encodes 393 amino acid residues and is one of the most commonly mutated genes in African-Asian populations (5). The p53 protein is a transcriptional product of TP53 that is associated with the inhibition of tumor cell division, induction of tumor cell apoptosis and the repair of DNA damage (6). In different types of tumors, mutations in the TP53 gene directly result the inactivation of p53 protein (4,6,7). TP53 mutations have been identified in malignant tumors of the lung, gastric, liver, breast and bladder, where p53 was found to be inactivated or dysfunctional (8,9). Therefore, it would be of great significance to utilize the tumor suppressive properties of functional p53 for use in cancer treatment.

Recombinant human adenovirus p53 (rAd-p53; also known as Gendicine®) is a replication-incompetent recombinant human serotype 5 adenovirus, where the virulent E1 region has been replaced by the human wild-type p53 expression cassette (10). It is the first commercially available product used for gene therapy and has been applied in the treatment of a number of cancer types, including head and neck cancer, epithelial ovarian carcinoma and liver cancer (11,12). rAd-p53 has been previously reported to enhance the sensitivity of gastric cancer cells to chemotherapy by regulating the expression of proteins that are associated with apoptosis (13). Clinical research has also demonstrated that rAd-p53-based transarterial chemoembolization is an effective and safe strategy for the treatment of unresectable liver cancer (14). However, treatment with rAd-p53 alone has proven to be insufficient for improving the survival of patients with cancer (15-17).

In some cancer malignancies, including in liver cancer, the aberrant tumor microenvironment may lead to subsequent mutation of the TP53 gene. In liver cancer cells, DNA damage and TP53 mutation have been previously associated with certain stimuli, including chronic inflammation and intracellular oxygen or nitrogen metabolites (18). Therefore, the introduction of exogenous wild-type TP53 in combination with anti-inflammatory and anti-oxidant agents that interfere with the tumor microenvironment but do not effect normal healthy cells, may produce improved therapeutic outcomes. Curcumin, a naturally occurring active compound extracted from the rhizome and root of the Curcuma longa plant, possesses antioxidant and anti-inflammatory properties (19) and has been demonstrated to be safe under clinical settings (20). As a result, curcumin has been considered as a promising therapeutic and preventative agent against liver cancer (21-23). However, the use of curcumin remains limited by its low bioavailability (21). In the present study, the treatment strategy of rAd-p53 combined with curcumin was investigated on HepG2 cells. Cell proliferation, apoptosis, expression of proteins targeted by the TP53 gene and the activation of mitogen-activated protein kinase (MAPK) signaling pathways were evaluated following treatment with rAd-p53 or curcumin individually and in combination. The results from the present study may assist in the development of therapeutic strategies involving rAd-p53 for use in the treatment of liver cancer.

Materials and methods

Cell culture and treatment

The human hepatocyte cell line HHL-5 and liver cancer cell lines HepG2, Hep3B and Huh-7 were supplied by the Type Culture Collection of the Chinese Academy of Sciences. Authentication of the cell lines was performed by STR profiling. Cells were maintained in RPMI 1640 medium (Hyclone; GE Healthcare Life Sciences) supplemented with 10% FBS (Gibco; Thermo Fisher Scientific, Inc.) at 37˚C under 5% CO2. All cells were subjected to treatment once ~90% confluence was achieved. Curcumin (Sigma-Aldrich; Merck KGaA) was dissolved in DMSO and administered at 37˚C to the cells at 10 µM. rAd-p53 (Sibiono GeneTech Co. Ltd.) was stored at -20˚C at a density of 1x1012 virus particles/ml. Prior to each experiment, cells were infected at 37˚C with rAd-p53 particles at a multiplicity of infection of 100 using a previously reported protocol (10). HHL-5 cells either remained untreated (control/Con) or were treated with curcumin (Cur) to evaluate the toxicity of curcumin on normal hepatocytes. HepG2, Hep3B and Huh-7 cells were divided into four groups in accordance with the treatments they Received: Con (control), rAd-p53 alone, Cur alone and Cur + rAd-p53.

Cell counting Kit-8 (CCK-8) assay

HHL-5 (untreated or treated with curcumin), HepG2, Hep3B and Huh-7 cells (either treated with/without rAD-p3 and/or curcumin) were seeded at the density of 3x103 cells/well into 96-well plates and subsequently cultured for 24, 48 and 72 h. Subsequently, a total of 10 µl CCK-8 solution (Bioswamp; Wuhan Beinglay Biotech Co., Ltd.) was added to each well, followed by further incubation at 37˚C for 4 h. Subsequently, a SpectraMax® 190 Microplate Reader (Molecular Devices, LLC) was used to measure the absorbance in each well at 450 nm.

Following treatment with or without rAD-p53 and/or curcumin for 72 h, HepG2 cells were incubated in RPMI-1640 medium containing 10% FBS without rAD-p53 and/or curcumin at 37˚C for 0, 24 and 48 h. Cell viability was detected using a CCK-8 assay as aforementioned. All experiments were performed in triplicate.

Wound healing assay

HepG2 cells were first seeded into six-well plates at 1x106 cells/well and incubated in DMEM (Hyclone; GE Healthcare Life Sciences) supplemented with 10% FBS. When ~90% confluence was reached, the cell monolayers were wounded by scratching with a sterile 200 μl plastic pipette tip, following which the cells were cultured in serum-free medium (DMEM) at 37˚C under 5% CO2 for 24, 48 or 72 h. The wounds were imaged using an inverted fluorescence microscope equipped with a camera (Nikon Corporation).

Flow cytometry analysis

Flow cytometry was performed to evaluate the apoptosis and cell cycle progression in HepG2 cells. For apoptosis, the Annexin V/PI staining method was performed according to the manufacturer's protocol (Bioswamp; Wuhan Beinglay Biotech Co., Ltd.). Following treatment, the cells (2x106 cells/ml) were digested using ethylenediaminetetraacetic acid-trypsin (Bioswamp; Wuhan Beinglay Biological Technology Co., Ltd.), washed with pre-cooled PBS and resuspended in binding buffer. Annexin V-fluorescein isothiocyanate and PI (10 µl each) were subsequently added to the cells, following which they were incubated for 30 min at 4˚C in the dark and subjected to the flow cytometry (Beckman Corporation) and the data were analyzed using CXP Analysis 2.0 software (Beckman Corporation).

For analyzing cell cycle progression, the harvested cells (2x107 cells/ml) were washed twice with pre-cooled PBS and incubated with a mixture of 100 µl 1 mg/ml RNase A (Takara Biotechnology Co., Ltd.) and 400 µl 50 µg/ml PI in the dark at room temperature for 10 min. The treated cells were then subjected to flow cytometry (Beckman Coulter, Inc.) and analyzed using ModFit LT 2.0 (Verity Software House).

Western blot analysis

The expression of proteins associated with cell cycle progression, apoptosis, TP53 targets p53 and p21, MAPK signaling and epithelial-mesenchymal transition (EMT) was evaluated using western blot analysis. Total protein content in HepG2 cells was extracted using a radioimmunoprecipitation assay lysis buffer (Bioswamp; Wuhan Beinglay Biotech Co., Ltd.) containing protease and phosphatase inhibitors. Proteins were quantified using the BCA kit (Bioswamp; Wuhan Beinglay Biotech Co., Ltd.). A total of 10 µg proteins were separated using SDS-PAGE (12%) and transferred onto PVDF (EMD Millipore). The membranes were then blocked with 5% skimmed milk for 2 h at room temperature and incubated with primary antibodies overnight at 4˚C. After washing, the membranes were incubated with secondary antibody for 1 h at room temperature. Immunoreactivity was visualized by colorimetric reaction using ECL substrate buffer (EMD Millipore). The membranes were then detected by an automatic chemiluminescence analyzer (Tanon-5200; Tanon Science and Technology Co., Ltd.) and the band gray values were read using TANON GIS 4.2 software (Tanon Science and Technology Co., Ltd.). All experiments were performed in triplicate. Detailed information of all antibodies used in the present study are presented in Table I.

Table I

Antibodies used in this study.

Table I

Antibodies used in this study.

AntibodySpeciesCompanyProduct codeDilutionProtein size (kDa)
Primary antibodies
     p53rabbitAbcamab1314421:1,00053
     p21rabbitAbcamab1091991:1,00018
     p-ERK1/2rabbitAbcamab2235001:40042-44
     ERK1/2rabbitAbcamab179421:1,00042-44
     p-p38MAPKrabbitAbcamab473631:1,00041
     p38MAPKrabbitAbcamab279861:1,00041
     p-JNKrabbitAbcamab1249561:5,00046-54
     JNKrabbitAbcamab1794611:1,00046-54
     Caspase3rabbitAbcamab904371:1,00032
     Caspase8rabbitAbcamab2274301:2,00055
     Caspase9rabbitAbcamab20131:2,00046
     BaxrabbitAbcamab531541:1,00021
     Bcl-2rabbitAbcamab1964951:2,00026
     Cyclin ArabbitAbcamab1377691:2,00049
     Cyclin ErabbitAbcamab339111:2,00050
     N-cadherinrabbitAbcamab182031:1,000100
     SnailrabbitAbcamab2163471:1,00029
     TwistrabbitAbcamab492541:40021
     GAPDHrabbitProteintech10494-1-AP1:5,00036
Secondary antibody
     Goat anti-rabbit IgGgoatBioswampSAB436581:20,000N/A
Reverse transcription-quantitative PCR (RT-qPCR)

p53 and p21 mRNA expression in HepG2 cells was measured using RT-qPCR. Total RNA was extracted using TRIzol® (Ambion; Thermo Fisher Scientific, Inc.) according to manufacturer's protocol, and reverse-transcribed into first-strand cDNA using the M-MLV kit (Takara Biotechnology Co., Ltd.) according to manufacturer's protocol. The temperature protocol were as follows: 42˚C for 1 h; 70˚C for 15 min and hold at 16˚C. The cDNA was subsequently used for qPCR using the SYBR® Green PCR kit (KAPA Biosystems; Roche diagnostics) according to manufacturer's protocols. The following thermocycling conditions were used for the PCR: 95˚C for 3 min; 39 cycles of denaturation at 95˚C for 5 sec, annealing at 56˚C for 10 sec and extension at 72˚C for 25 sec and final extension at 65˚C for 5 sec and 95˚C for 50 sec. The primer sequences used are as follows: p53 forward, 5'-ATGTTTGTGCCTG CCT-3' and reverse, 5'-CAGTGGTTTCTTCTTTGG-3'; p21 forward, 5'-CGTGAGCGATGGAACTT-3' and reverse, 5'-GCAGAGCAGGTGAGGTG-3' and GAPDH forward, 5'-CCACTCCTCCACCTTTG-3' and reverse, 5'-CACCAC CCTGTTGCTGT-3'. The data were obtained using QuantStudio™ 6 Flex Real-Time PCR System (Applied Biosystems; Thermo Fisher Scientific, Inc.) and analyzed with the 2-ΔΔCq method (24). The expression of all mRNA was normalized to that of GAPDH. All experiments were performed in triplicate.

Statistical analysis

The data are presented as the mean ± standard deviation and analyzed using SPSS 19 (IBM Corp.). Differences between ≥2 groups were analyzed using one-way ANOVA followed by a least significant difference whereas those between two groups were analyzed using an unpaired t-test. P<0.05 was considered to indicate a statistically significant difference.

Results

Cytotoxicity effect of rAd-p53 and/or curcumin on HepG2, Hep3B and Huh-7 cells

The extent of curcumin cytotoxicity was measured in HHL-5 cells. Following 72 h curcumin treatment (10 µM), cell viability was ~70% of that in control cells (Fig. 1A). rAd-p53 and curcumin treatments alone reduced the cell viability of HepG2, Hep3B and Huh-7 cells, whilst the combined administration of rAd-p53 and curcumin produced additive inhibitory effects compared with Cur, in a time-dependent manner (Fig. 1B-D). Since HepG2 cells appeared to exhibit the highest sensitivity to curcumin and/or rAd-p53 among the liver cancer cell lines (Fig. 1D), it was selected for subsequent experiments. HepG2 cell viability continued to decrease up to 48 h after rAD-p53 and/or curcumin was removed (Fig. 1E). These results demonstrated that the combined administration of curcumin and rAd-p53 synergistically reduced HepG2, Hep3B and Huh-7 cell viability.

Combined effect of rAd-p53 and curcumin treatment on HepG2 EMT

The wound healing ability of HepG2 cells treated with either rAd-p53 or curcumin appeared to be inferior compared with that observed for non-treated cells; with the combined treatment of the two agents potentiating this inhibition further in a time-dependent manner (Fig. 2A). The expression of proteins associated with EMT were then evaluated using western blot analysis. Compared with control cells, cells treated with either rAd-p53 or Cur exhibited reduced N-cadherin, snail and twist expression, which was reduced further following combined rAd-p53 and curcumin treatment (Fig. 2B). These observations indicated that the combined administration of curcumin and rAd-p53 additively suppressed EMT in HepG2 cells in a time dependent manner.

Combined effect of rAd-p53 and curcumin administration on HepG2 apoptosis and intracellular protein expression

The apoptosis of HepG2 cells following a number of treatments is presented in Fig. 3A. The percentage of apoptotic cells in the control group was revealed to be ~2.19%, which were increased to 12.76 and 15.47% following the individual treatment of either rAd-p53 or Cur alone after 24 h, respectively (Fig. 3A). The combined administration of rAd-p53 and curcumin resulted in a further increase in the percentage of apoptotic cells to 20.29%. After 72 h, whilst the percentage of apoptotic cells in the control group increased slightly (5.85%), those in the treatment groups were more prominent. A total of 45.61% of apoptotic cells were observed in the Cur + rAd-p53 group (Fig. 3A).

The expression of proteins associated with apoptosis in HepG2 cells was subsequently evaluated using western blot analysis. Compared with control cells, cells treated with rAd-p53 alone demonstrated significantly higher expression of pro-apoptotic proteins Bax and caspases 3, 8 and 9, which were potentiated further in cells treated with rAd-p53 and curcumin together (Fig. 4). Following the same rAd-p53 and/or Cur treatment regimens, the expression of the anti-apoptotic protein Bcl-2 exhibited the opposite trend compared with that of the pro-apoptotic proteins (Fig. 4). These results were supported by those obtained from the Annexin V/PI assay. The results collectively indicated that the combined administration of curcumin and rAd-p53 enhanced HepG2 apoptosis.

Combined effect of rAd-p53 and curcumin on HepG2 cell cycle progression and the expression of associated proteins

Treatment with either Cur or rAd-p53 reduced the proportion of cells in the G1/S phase whilst increasing those in the G2/M phase compared with the control HepG2 cells. This effect was potentiated further in cells treated with rAd-p53 and curcumin combined (Fig. 3B). No notable differences were observed in the proportion of cells in S phase between all four treatment groups (Fig. 3B). Supporting this observation, the expression of Cyclins A and E, which are proteins associated with cell cycle progression, were significantly increased by either rAd-p53 (Fig. 4). This increase was potentiated further following the combined administration of rAd-p53 and curcumin (Fig. 4). These results indicated that the combined administration of curcumin and rAd-p53 induced a stronger effect compared with Cur treatment alone in altering cell cycle progression.

Combined effect of rAd-p53 and curcumin on the expression of p53, p21 and MAPKs

In all time points tested, the administration of rAd-p53 alone significantly upregulated p53 and p21 expression in HepG2 cells, which was potentiated further by combined rAd-p53 and curcumin treatment (Fig. 4). Similar trends were observed for the phosphorylation levels of p38 MAPK (p-p38 MAPK) and c-Jun N-terminal kinase (p-JNK), whereas that of extracellular signal-regulated kinases (p-ERK1/2) exhibited the opposite trend (Fig. 5). These results demonstrated that the combined administration of rAd-p53 and curcumin exerted additive regulatory effects on associated signaling pathways compared with Cur treatment alone.

Discussion

TP53 is an important tumor suppressor gene in the human body, the expression of which is reduced in cancer cells (21). TP53 is found to be functionally inactivated in 50% of all human cancer cases and 61% of all liver cancer human cases (25,26), and the downregulation of p53 protein expression promotes the development of liver cancer (27). Consequently, TP53 is a candidate gene for gene-targeted therapy in human malignancies (28). rAd-p53 is the first commercially available product for gene therapy that has been applied in the treatment of head and neck cancer, epithelial ovarian carcinoma and liver cancer (11,12). In most cases, liver cancer occurs and develops as a result of inflammation and oxidative stress (18,21). In the present study, the therapeutic effects of combining rAd-p53 with curcumin, which is a compound exhibiting excellent anti-inflammatory and antioxidant properties (20,21), was explored in liver cancer, using HepG2 cells as the model cell line. This combinatorial administration was demonstrated to synergistically promote apoptosis, inhibit G2/M phase progression and suppress EMT in HepG2 cells.

rAd-p53 is a relatively effective and safe means of treatment for liver cancer (29,30) and is usually applied in combination with other therapies, including transarterial chemoembolization (14) and 5-fluorouracil administration (31). Previous clinical studies have revealed that rAd-p53 injection may improve the survival rate of patients with liver cancer (14,32,33). Additionally, the combination of rAd-p53 with N-Myc downstream-regulated gene 2 increased p53-mediated apoptosis of HepG2 and Huh7 cells in a previous study (34). In the present study, rAd-p53 was revealed to upregulate the expression of p53 in HepG2 cells compared with non-treated cells. This observation is consistent with a previous report, where the intratumoral injection of rAd-p53 resulted in increased p53 expression in prostate cancer (35,36). Downstream, the expression of the p53-targeted gene CDKN1 followed a trend parallel to that of TP53, suggesting that the p53 protein produced following rAd-p53 treatment is physiologically active. The apoptosis rate was indicated to be markedly increased, as demonstrated by the upregulation of the pro-apoptotic proteins caspases 3, 8 and 9 in addition to Bax and coupled with the downregulation of the anti-apoptotic protein Bcl-2. EMT was also notably suppressed, as demonstrated by the downregulation of N-cadherin, snail and twist expression, which are well documented markers of EMT (37,38).

TP53 often serves as a ‘guardian of the genome’, the deletion of which may result in the uncontrolled proliferation of tumor cells (39). The upregulation of TP53 expression that is induced by rAd-p53 treatment promoted human cervical cancer cell apoptosis through activation of the Bax gene and suppression of the Bcl-x gene and resulted in cell cycle arrest at the G2/M phase (40). The results of the present study are consistent with those observed in previous reports, which have demonstrated that TP53 activation is associated with liver cancer cell apoptosis by regulating the expression of Bcl-2 and caspases (41,42), in addition to inhibiting cancer cell migration (43-45). TP53 activation has also been previously reported to serve an inhibitory role in the EMT process, in human oral mucosal fibroblasts and oral submucous fibrosis by downregulating N-cadherin expression (46) and in colorectal cancer cells by downregulating Snail expression (37), which are findings consistent with the results of the present study.

Curcumin possesses anti-inflammatory and antioxidant properties and has also been observed to upregulate TP53 expression in tumor cells to exert several therapeutic effects (47,48). Curcumin induces apoptosis and cell cycle arrest of cancer cells by targeting regulatory p53(49). Previous in vivo and in vitro experiments have demonstrated that curcumin in combination with metformin induces apoptosis and suppresses the proliferation, invasion and metastasis of liver cancer cells by upregulating TP53 (50). In the present study, the combination of rAd-p53 and curcumin led to a higher expression of p53 compared with Cur treatment alone, synergistically promoting apoptosis, inhibiting cell proliferation and migration by regulating TP53. In addition, curcumin exerted anti-tumor effects by regulating the MAPK pathways. There are three subfamilies of MAPKs, including p38MAPK, JNKs and ERKs, all of which are related to apoptosis (51). A previous report has suggested that curcumin treatment induced retinoblastoma cell apoptosis by activating p38 MAPK and JNK (52). Similarly, curcumin-induced p38 MAPK activation resulted in FasL-associated apoptosis in human hepatocellular carcinoma Huh7 cells (53). Curcumin also induced apoptosis in HepG2 cells by activating the ROS-ASK1-JNK pathway (54). Consistent with previous studies, the co-treatment of rAd-p53 with curcumin in the present study resulted in the additive potentiation of p38MAPK and JNK activation, potentially resulting in apoptosis in this manner.

In conclusion, rAd-p53 and curcumin were applied individually or in combination to explore their influence on the liver cancer cell line HepG2. Compared with Cur treatment alone, the combined treatment synergistically promoted liver cancer apoptosis and inhibited cell migration. Mechanistically, these observed effects may be associated with TP53 expression and subsequent MAPK signaling. Overall, the present study provides new insights into possible targets for effective liver cancer therapy.

Acknowledgements

Not applicable.

Funding

No funding was received.

Availability of data and materials

The datasets used and/or analyzed during the current study are available from the corresponding author on reasonable request.

Authors' contributions

JQ and JY participated in the design of this work. JQ, WL, MC, WG, CZ and BG performed the experiments and analyzed data. JQ drafted the manuscript and JY revised the manuscript. All authors read and approved the final manuscript.

Ethics approval and consent to participate

Not applicable.

Patient consent for publication

Not applicable.

Competing interests

The authors declare that they have no competing interests.

References

1 

Yu LX and Schwabe RF: The gut microbiome and liver cancer: Mechanisms and clinical translation. Nat Rev Gastroenterol Hepatol. 14:527–539. 2017.PubMed/NCBI View Article : Google Scholar

2 

Jiang JF, Lao YC, Yuan BH, Yin J, Liu X, Chen L and Zhong JH: Treatment of hepatocellular carcinoma with portal vein tumor thrombus: Advances and challenges. Oncotarget. 8:33911–33921. 2017.PubMed/NCBI View Article : Google Scholar

3 

Bruix J, Han KH, Gores G, Llovet JM and Mazzaferro V: Liver cancer: Approaching a personalized care. J Hepatol. 62:S144–S156. 2015.PubMed/NCBI View Article : Google Scholar

4 

Poulain S, Roumier C, Bertrand E, Renneville A, Caillault-Venet A, Doye E, Geffroy S, Sebda S, Nibourel O, Nudel M, et al: TP53 mutation and its prognostic significance in Waldenstrom's macroglobulinemia. Clin Cancer Res. 23:6325–6335. 2017.PubMed/NCBI View Article : Google Scholar

5 

Friemel J, Rechsteiner M, Bawohl M, Frick L, Müllhaupt B, Lesurtel M and Weber A: Liver cancer with concomitant TP53 and CTNNB1 mutations: A case report. BMC Clin Pathol. 16(7)2016.PubMed/NCBI View Article : Google Scholar

6 

Vogelstein B, Lane D and Levine AJ: Surfing the p53 network. Nature. 408:307–310. 2000.PubMed/NCBI View Article : Google Scholar

7 

McCubrey JA, Lertpiriyapong K, Fitzgerald TL, Martelli AM, Cocco L, Rakus D, Gizak A, Libra M, Cervello M, Montalto G, et al: Roles of TP53 in determining therapeutic sensitivity, growth, cellular senescence, invasion and metastasis. Adv Biol Regul. 63:32–48. 2017.PubMed/NCBI View Article : Google Scholar

8 

Soussi T and Wiman KG: TP53: An oncogene in disguise. Cell Death Differ. 22:1239–1249. 2015.PubMed/NCBI View Article : Google Scholar

9 

Kashofer K and Regauer S: Analysis of full coding sequence of the TP53 gene in invasive vulvar cancers: Implications for therapy. Gynecol Oncol. 146:314–318. 2017.PubMed/NCBI View Article : Google Scholar

10 

Li J, Pan J, Zhu X, Su Y, Bao L, Qiu S, Zou C, Cai Y, Wu J and Tham IW: Recombinant adenovirus-p53 (Gendicine) sensitizes a pancreatic carcinoma cell line to radiation. Chin J Cancer Res. 25:715–721. 2013.PubMed/NCBI View Article : Google Scholar

11 

Räty JK, Pikkarainen JT, Wirth T and Ylä-Herttuala S: Gene therapy: The first approved gene-based medicines, molecular mechanisms and clinical indications. Curr Mol Pharmacol. 1:13–23. 2008.PubMed/NCBI View Article : Google Scholar

12 

Li Y, Li B, Li CJ and Li LJ: Key points of basic theories and clinical practice in rAd-p53 (Gendicine ™) gene therapy for solid malignant tumors. Expert Opin Biol Ther. 15:437–454. 2015.PubMed/NCBI View Article : Google Scholar

13 

Chen GX, Zheng LH, Liu SY and He XH: rAd-p53 enhances the sensitivity of human gastric cancer cells to chemotherapy. World J Gastroenterol. 17:4289–4297. 2011.PubMed/NCBI View Article : Google Scholar

14 

Shen A, Liu S, Yu W, Deng H and Li Q: p53 gene therapy-based transarterial chemoembolization for unresectable hepatocellular carcinoma: A prospective cohort study. J Gastroenterol Hepatol. 30:1651–1656. 2015.PubMed/NCBI View Article : Google Scholar

15 

Li Y, Li LJ, Wang LJ, Zhang Z, Gao N, Liang CY, Huang YD and Han B: Selective intra-arterial infusion of rAd-p53 with chemotherapy for advanced oral cancer: A randomized clinical trial. BMC Med. 12(16)2014.PubMed/NCBI View Article : Google Scholar

16 

Guan YS, Liu Y, Zou Q, He Q, La Z, Yang L and Hu Y: Adenovirus-mediated wild-type p53 gene transfer in combination with bronchial arterial infusion for treatment of advanced non-small-cell lung cancer, one year follow-up. J Zhejiang Univ Sci B. 10:331–340. 2009.PubMed/NCBI View Article : Google Scholar

17 

Buller RE, Runnebaum IB, Karlan BY, Horowitz JA, Shahin M, Buekers T, Petrauskas S, Kreienberg R, Slamon D and Pegram M: A phase I/II trial of rAd/p53 (SCH 58500) gene replacement in recurrent ovarian cancer. Cancer Gene Ther. 9:553–566. 2002.PubMed/NCBI View Article : Google Scholar

18 

Staib F, Hussain SP, Hofseth LJ, Wang XW and Harris CC: TP53 and liver carcinogenesis. Hum Mutat. 21:201–216. 2003.PubMed/NCBI View Article : Google Scholar

19 

Chin KY: The spice for joint inflammation: Anti-inflammatory role of curcumin in treating osteoarthritis. Drug Des Devel Ther. 10:3029–3042. 2016.PubMed/NCBI View Article : Google Scholar

20 

Lestari ML and Indrayanto G: Curcumin. Profiles Drug Subst Excip Relat Methodol. 39:113–204. 2014.PubMed/NCBI View Article : Google Scholar

21 

Darvesh AS, Aggarwal BB and Bishayee A: Curcumin and liver cancer: A review. Curr Pharm Biotechnol. 13:218–228. 2012.PubMed/NCBI View Article : Google Scholar

22 

El-Houseini ME, El-Agoza IA, Sakr MM and El-Malky GM: Novel protective role of curcumin and taurine combination against experimental hepatocarcinogenesis. Exp Ther Med. 13:29–36. 2017.PubMed/NCBI View Article : Google Scholar

23 

Marquardt JU, Gomez-Quiroz L, Arreguin Camacho LO, Pinna F, Lee YH, Kitade M, Domínguez MP, Castven D, Breuhahn K, Conner EA, et al: Curcumin effectively inhibits oncogenic NF-κB signaling and restrains stemness features in liver cancer. J Hepatol. 63:661–669. 2015.PubMed/NCBI View Article : Google Scholar

24 

Livak KJ and Schmittgen TD: Analysis of relative gene expression data using real-time quantitative PCR and the 2(-ΔΔC(T)) method. Methods. 25:402–408. 2001.PubMed/NCBI View Article : Google Scholar

25 

Hsia CC, Nakashima Y, Thorgeirsson SS, Harris CC, Minemura M, Momosaki S, Wang NJ and Tabor E: Correlation of immunohistochemical staining and mutations of p53 in human hepatocellular carcinoma. Oncol Rep. 7:353–356. 2000.PubMed/NCBI

26 

Honda K, Sbisà E, Tullo A, Papeo PA, Saccone C, Poole S, Pignatelli M, Mitry RR, Ding S, Isla A, et al: p53 mutation is a poor prognostic indicator for survival in patients with hepatocellular carcinoma undergoing surgical tumour ablation. Br J Cancer. 77:776–782. 1998.PubMed/NCBI View Article : Google Scholar

27 

Wu W, Liu S, Liang Y, Zhou Z, Bian W and Liu X: Stress hormone cortisol enhances Bcl2 like-12 expression to inhibit p53 in hepatocellular carcinoma cells. Dig Dis Sci. 62:3495–3500. 2017.PubMed/NCBI View Article : Google Scholar

28 

Li VD, Li KH and Li JT: TP53 mutations as potential prognostic markers for specific cancers: Analysis of data from The Cancer Genome Atlas and the International Agency for Research on Cancer TP53 Database. J Cancer Res Clin Oncol. 145:625–636. 2019.PubMed/NCBI View Article : Google Scholar

29 

Tu K, Zheng X, Zhou Z, Li C, Zhang J, Gao J, Yao Y and Liu Q: Recombinant human adenovirus-p53 injection induced apoptosis in hepatocellular carcinoma cell lines mediated by p53-Fbxw7 pathway, which controls c-Myc and cyclin E. PLoS One. 8(e68574)2013.PubMed/NCBI View Article : Google Scholar

30 

Chen SX, Xu WD, Yin GW, Xi W, Chen J, Xu QY and Ma GJ: Clinical therapeutic effect and biological monitoring of p53 gene in advanced hepatocellular carcinoma. Zhonghua Yi Xue Za Zhi. 90:2182–2186. 2010.PubMed/NCBI(In Chinese).

31 

Tian G, Liu J, Zhou JS and Chen W: Multiple hepatic arterial injections of recombinant adenovirus p53 and 5-fluorouracil after transcatheter arterial chemoembolization for unresectable hepatocellular carcinoma: A pilot phase II trial. Anticancer Drugs. 20:389–395. 2009.PubMed/NCBI View Article : Google Scholar

32 

Guan YS, Liu Y, He Q, Li X, Yang L, Hu Y and La Z: p53 gene therapy in combination with transcatheter arterial chemoembolization for HCC: One-year follow-up. World J Gastroenterol. 17:2143–2149. 2011.PubMed/NCBI View Article : Google Scholar

33 

Yang ZX, Wang D, Wang G, Zhang QH, Liu JM, Peng P and Liu XH: Clinical study of recombinant adenovirus-p53 combined with fractionated stereotactic radiotherapy for hepatocellular carcinoma. J Cancer Res Clin Oncol. 136:625–630. 2010.PubMed/NCBI View Article : Google Scholar

34 

Cao W, Zhang JL, Feng DY, Liu XW, Li Y, Wang LF, Yao LB, Zhang H and Zhang J: The effect of adenovirus-conjugated NDRG2 on p53-mediated apoptosis of hepatocarcinoma cells through attenuation of nucleotide excision repair capacity. Biomaterials. 35:993–1003. 2014.PubMed/NCBI View Article : Google Scholar

35 

Sasaki R, Shirakawa T, Zhang ZJ, Tamekane A, Matsumoto A, Sugimura K, Matsuo M, Kamidono S and Gotoh A: Additional gene therapy with Ad5CMV-p53 enhanced the efficacy of radiotherapy in human prostate cancer cells. Int J Radiat Oncol Biol Phys. 51:1336–1345. 2001.PubMed/NCBI View Article : Google Scholar

36 

Shirakawa T, Gotoh A, Gardner TA, Kao C, Zhang ZJ, Matsubara S, Wada Y, Hinata N, Fujisawa M, Hanioka K, et al: p53 adenoviral vector (Ad-CMV-p53) induced prostatic growth inhibition of primary cultures of human prostate and an experimental rat model. J Gene Med. 2:426–432. 2000.PubMed/NCBI View Article : Google Scholar

37 

Bai Z, Wang J, Wang T, Li Y, Zhao X, Wu G, Yang Y, Deng W and Zhang Z: The miR-495/Annexin A3/P53 axis inhibits the invasion and EMT of colorectal cancer cells. Cell Physiol Biochem. 44:1882–1895. 2017.PubMed/NCBI View Article : Google Scholar

38 

Angadi PV, Patil PV, Angadi V, Mane D, Shekar S, Hallikerimath S, Kale AD and Kardesai SG: Immunoexpression of epithelial mesenchymal transition proteins E-Cadherin, β-Catenin, and N-Cadherin in oral squamous cell carcinoma. Int J Surg Pathol. 24:696–703. 2016.PubMed/NCBI View Article : Google Scholar

39 

Curiel DT, Gerritsen WR and Krul MR: Progress in cancer gene therapy. Cancer Gene Ther. 7:1197–1199. 2000.PubMed/NCBI View Article : Google Scholar

40 

Liu YG, Zheng XL and Liu FM: The mechanism and inhibitory effect of recombinant human P53 adenovirus injection combined with paclitaxel on human cervical cancer cell HeLa. Eur Rev Med Pharmacol Sci. 19:1037–1042. 2015.PubMed/NCBI

41 

Liao W, Liu J, Liu B, Huang X, Yin Y, Cai D, Li M and Zhu R: JIB 04 induces cell apoptosis via activation of the p53/Bcl 2/caspase pathway in MHCC97H and HepG2 cells. Oncol Rep. 40:3812–3820. 2018.PubMed/NCBI View Article : Google Scholar

42 

Cao Y, Cao J, Yu B, Wang S, Liu L, Tao L and Sun W: Berbamine induces SMMC-7721 cell apoptosis via upregulating p53, downregulating survivin expression and activating mitochondria signaling pathway. Exp Ther Med. 15:1894–1901. 2018.PubMed/NCBI View Article : Google Scholar

43 

Tamura M, Sasaki Y, Koyama R, Takeda K, Idogawa M and Tokino T: Forkhead transcription factor FOXF1 is a novel target gene of the p53 family and regulates cancer cell migration and invasiveness. Oncogene. 33:4837–4846. 2014.PubMed/NCBI View Article : Google Scholar

44 

Liu Y, Li L, Liu Y, Geng P, Li G, Yang Y and Song H: RECK inhibits cervical cancer cell migration and invasion by promoting p53 signaling pathway. J Cell Biochem. 119:3058–3066. 2018.PubMed/NCBI View Article : Google Scholar

45 

Zhang B, Yin X and Sui S: Resveratrol inhibited the progression of human hepatocellular carcinoma by inducing autophagy via regulating p53 and the phosphoinositide 3 kinase/protein kinase B pathway. Oncol Rep. 40:2758–2765. 2018.PubMed/NCBI View Article : Google Scholar

46 

Zheng L, Guan ZJ, Pan WT, Du TF, Zhai YJ and Guo J: Tanshinone suppresses arecoline-induced epithelial-mesenchymal transition in oral submucous fibrosis by epigenetically reactivating the p53 pathway. Oncol Res. 26:483–494. 2018.PubMed/NCBI View Article : Google Scholar

47 

Sidhar H and Giri RK: Induction of Bex genes by curcumin is associated with apoptosis and activation of p53 in N2a neuroblastoma cells. Sci Rep. 7(41420)2017.PubMed/NCBI View Article : Google Scholar

48 

Li W, Wang Y, Song Y, Xu L, Zhao J and Fang B: A preliminary study of the effect of curcumin on the expression of p53 protein in a human multiple myeloma cell line. Oncol Lett. 9:1719–1724. 2015.PubMed/NCBI View Article : Google Scholar

49 

Kasi PD, Tamilselvam R, Skalicka-Woźniak K, Nabavi SF, Daglia M, Bishayee A, Pazoki-Toroudi H and Nabavi SM: Molecular targets of curcumin for cancer therapy: An updated review. Tumour Biol. 37:13017–13028. 2016.PubMed/NCBI View Article : Google Scholar

50 

Zhang HH, Zhang Y, Cheng YN, Gong FL, Cao ZQ, Yu LG and Guo XL: Metformin incombination with curcumin inhibits the growth, metastasis, and angiogenesis of hepatocellular carcinoma in vitro and in vivo. Mol Carcinog. 57:44–56. 2018.PubMed/NCBI View Article : Google Scholar

51 

Wada T and Penninger JM: Mitogen-activated protein kinases in apoptosis regulation. Oncogene. 23:2838–2849. 2004.PubMed/NCBI View Article : Google Scholar

52 

Yu X, Zhong J, Yan L, Li J, Wang H, Wen Y and Zhao Y: Curcumin exerts antitumor effects in retinoblastoma cells by regulating the JNK and p38 MAPK pathways. Int J Mol Med. 38:861–868. 2016.PubMed/NCBI View Article : Google Scholar

53 

Wang WZ, Li L, Liu MY, Jin XB, Mao JW, Pu QH, Meng MJ, Chen XG and Zhu JY: Curcumin induces FasL-related apoptosis through p38 activation in human hepatocellular carcinoma Huh7 cells. Life Sci. 92:352–358. 2013.PubMed/NCBI View Article : Google Scholar

54 

Zheng R, You Z, Jia J, Lin S, Han S, Liu A, Long H and Wang S: Curcumin enhances the antitumor effect of ABT-737 via activation of the ROS-ASK1-JNK pathway in hepatocellular carcinoma cells. Mol Med Rep. 13:1570–1576. 2016.PubMed/NCBI View Article : Google Scholar

Related Articles

Journal Cover

November-2020
Volume 20 Issue 5

Print ISSN: 1792-0981
Online ISSN:1792-1015

Sign up for eToc alerts

Recommend to Library

Copy and paste a formatted citation
x
Spandidos Publications style
Qu J, Lu W, Chen M, Gao W, Zhang C, Guo B and Yang J: Combined effect of recombinant human adenovirus p53 and curcumin in the treatment of liver cancer. Exp Ther Med 20: 18, 2020
APA
Qu, J., Lu, W., Chen, M., Gao, W., Zhang, C., Guo, B., & Yang, J. (2020). Combined effect of recombinant human adenovirus p53 and curcumin in the treatment of liver cancer. Experimental and Therapeutic Medicine, 20, 18. https://doi.org/10.3892/etm.2020.9145
MLA
Qu, J., Lu, W., Chen, M., Gao, W., Zhang, C., Guo, B., Yang, J."Combined effect of recombinant human adenovirus p53 and curcumin in the treatment of liver cancer". Experimental and Therapeutic Medicine 20.5 (2020): 18.
Chicago
Qu, J., Lu, W., Chen, M., Gao, W., Zhang, C., Guo, B., Yang, J."Combined effect of recombinant human adenovirus p53 and curcumin in the treatment of liver cancer". Experimental and Therapeutic Medicine 20, no. 5 (2020): 18. https://doi.org/10.3892/etm.2020.9145