Potential therapeutic value of dendritic cells loaded with NY‑ESO‑1 protein for the immunotherapy of advanced hepatocellular carcinoma

  • Authors:
    • Yuqing Chen
    • Aimin Huang
    • Meiqin Gao
    • Yongqin Yan
    • Wenmin Zhang
  • View Affiliations

  • Published online on: September 27, 2013     https://doi.org/10.3892/ijmm.2013.1510
  • Pages: 1366-1372
Metrics: Total Views: 0 (Spandidos Publications: | PMC Statistics: )
Total PDF Downloads: 0 (Spandidos Publications: | PMC Statistics: )


Abstract

NY‑ESO‑1 is one of the most immunogenic cancer-testis (CT) antigens. Cancer vaccine trials based on NY‑ESO‑1 are currently ongoing. Dendritic cells (DCs) are the most potent antigen-presenting cells. The immune functions of DCs in a number of tumors have been identified; however, the potential therapeutic value of DCs pulsed with NY‑ESO‑1 in hepatocellular carcinoma (HCC) has not been extensively investigated. The objectives of the present study were to evaluate T cell response following stimulation with DCs pulsed with the recombinant NY‑ESO‑1 protein (rESO) and to establish a correlation between NY‑ESO‑1 expression and clinicopathological features in HCC patients. DCs were generated with granulocyte/macrophage colony-stimulating factor (GM-CSF) and interleukin-4 (IL‑4) from human peripheral blood mononuclear cells. A mixed T cell reaction with DCs loaded with recombinant NY‑ESO‑1 protein (rESO-DCs) was evaluated by MTT assay. T cell responses against HCC cell lines were analyzed by measuring lactate dehydrogenase (LDH) activity. The protein levels of NY‑ESO‑1 were detected by immunohistochemistry (IHC) in a tissue microarray (TMA) containing 190 HCC samples. NY‑ESO‑1 transcript abundance was determined by reverse transcriptase-polymerase chain reaction (RT-PCR) in 54 out of the 190 HCC samples. The results revealed that mature DCs were induced and that rESO‑DCs significantly stimulated T cell proliferation. The specific lysis of T cells stimulated with rESO‑DCs was significantly higher in the NY‑ESO‑1-positive HCC cells compared with the NY‑ESO‑1-negative cells and the other controls (p<0.01). NY‑ESO‑1 was expressed in 15.8% (30/190)of the HCC samples, as shown by IHC and in 24.1% (13/54) of the samples, as shown by RT-PCR. The frequency of NY‑ESO‑1 expression was significantly higher in HCC patients with portal vein tumor thrombosis (24.6%) compared with those without thrombosis (11.2%, p=0.013). Our data suggest that DCs loaded with NY‑ESO‑1 protein stimulate antigen-specific T cell responses against HCC cells in vitro. NY‑ESO‑1 may thus be used as a potential target for immunotherapy in advanced HCC.

Introduction

Hepatocellular carcinoma (HCC) is the fifth most common type of cancer worldwide and is particularly prevalent in China (1,2). Currently, treatments for HCC include surgical resection, chemotherapy and liver transplantation (3). However, the outcomes remain dismal. The 5-year survival rate for patients with HCC has been reported to be 30–50% (4,5). Thus, novel, alternative therapeutic options for HCC are urgently required. Immunotherapy for cancer has received much attention in recent years. A number of tumor antigens, such as human telomerase-reverse transcriptase (hTERT) or alpha-fetoprotein (AFP) have been identified as immunotherapeutic targets, and a number of immunotherapeutic trials have been performed to evaluate the potential therapeutic value of HCC immunotherapy (6).

Cancer-testis (CT) antigens have been considered attractive targets for cancer immunotherapy due to their restricted expression patterns in a variety of tumors and normal tissues (7). To date, >150 genes or gene families encoding CT antigens have been identified (8). However, only a limited number of CT antigens have been shown to elicit both humoral and cellular responses. NY-ESO-1 (cancer/testis antigen 1B), also known as CTAG1, is one of the most immunogenic CT antigens (9). It was originally found in esophageal cancer by serological recombinant cDNA expression cloning (SEREX) (10) and is expressed in several tumors, including HCC (1114). NY-ESO-1 expression is associated with a poor tumor outcome and is recognized as a potential biomarker for the prediction of tumor recurrence and treatment outcomes in patients with gastrointestinal stromal tumors and cutaneous melanomas (15).

Due to its expression patterns and strong immunogenicity, NY-ESO-1 has emerged as one of the most attractive targets for cancer vaccines (7,9). The NY-ESO-1 vaccine, based on peptide or protein, has been tested in the treatment of patients with tumors expressing NY-ESO-1, including lung cancer, ovarian cancer, esophageal cancer and melanoma (1620). The majority of the patients showed enhanced immune responses and disease stabilization was achieved in some patients (20). Furthermore, these studies illustrated the safety of various preparations of NY-ESO-1 vaccines. The use of in vitro-generated, autologous dendritic cells (DCs) as a cellular adjuvant for vaccine delivery has been widely tested in cancer patients (21). The potential efficacy of induced immunity against HCC has been supported by a report that the immunization of 2 HCC patients with autologous HCC lysate-loaded DCs resulted in the prolonged survival of >3 years in 1 patient (22). However, whether DCs pulsed with NY-ESO-1 protein can induce antigen-specific immune responses against HCC remains unclear. In addition, the correlation between NY-ESO-1 expression and clinical parameters has not been extensively investigated.

In a previous study, we purified the recombinant NY-ESO-1 protein (rESO) (23). In this study we aimed to evaluate T cell response against HCC cell lines following incubation with DCs loaded with rESO. Furthermore, we assessed the mRNA and protein abundance of NY-ESO-1 in HCC samples and determined the correlation between NY-ESO-1 expression and clinical parameters.

Materials and methods

Patients and samples

A total of 190 paraffin-embedded HCC specimens and their adjacent non-cancerous tissues were collected at the Center for Liver Disease, the First Affiliated Hospital, Fujian Medical University, Fuzhou, China, between 2007 and 2009. Frozen tissues were available in 54 cases. They were frozen immediately in liquid nitrogen after removal from surgical resection and stored at −80°C until use. Informed consent was obtained from all patients. The clinicopathological parameters of these cases are summarized in Table I. Tissue microarrays (TMAs) of the HCC and adjacent non-cancerous liver samples were prepared according to standard procedures (Beecher Instruments Inc., Silver Spring, MD, USA). The study was approved by the Ethical Review Board of Fujian Medical University.

Table I

Clinicopathological characteristics of HCC patients.

Table I

Clinicopathological characteristics of HCC patients.

CharacteristicParaffin-embeded HCC specimensFresh-frozen HCC specimens
No. of patients19054
Average age (years)4950
Age range (years)21–7532–70
Male/female162/2844/10
HBsAg-positive15544
AFP (≥20 ng/ml)13239
With cirrhosis17248
With portal vein tumor thrombosis6518
Edmondson’s classification (grades I–II/III–IV)103/8729/25

[i] HBsAg, hepatitis B surface antigen; AFP, alpha-fetoprotein. HCC, hepatocellular carcinoma.

HCC cell lines

The HCC cell lines, H4M and H2P, were kindly provided by Dr JianMing Wen at the Department of Pathology, the First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China. HCC cells were cultured in RPMI-1640 medium with 10% fetal bovine serum (FBS), L-glutamine, penicillin (100 IU/ml) and streptomycin (100 μg/ml) at 37°C.

Preparation of DCs

Peripheral blood mononuclear cells (PBMCs) from healthy volunteers were isolated from blood by Ficoll-Hypaque density gradient centrifugation (Amersham Biosciences, Uppsala, Sweden). The cells were then seeded on 6-well plates for 2 h at a density of 2–3×106 cells/ml. Non-adherent cells were removed and adherent cells were incubated in RPMI-1640 medium supplemented with 20% FBS, 1,000 U/ml of granulocyte macrophage colony-stimulating factor (GM-CSF) and 500 U/ml of interleukin-4 (IL-4; PeproTech Inc., Rocky Hill, NJ, USA). After 3 days of incubation, the old medium with floating cells was gently removed and replaced with fresh medium. After 5 days of incubation, 1/3 of the cells were collected as immature DCs (imDCs). The remaining cells were treated with rESO or IL-4 at a concentration of 50 μg/ml for 24 h. The cells were then incubated with 10 ng/ml tumor necrosis factor-α (TNF-α) for 48 h to induce the formation of mature DCs (mDCs). ImDCs and mDCs were assayed by flow cytometry.

Detection of T cell response

T cells (4×105 cells/well) were incubated with imDCs or mDCs at a ratio of 20:1 at 37°C for 72 h in RPMI-1640 medium with 20% FBS, 100 U/ml interleukin-2 (IL-2) and 20 μg/ml phytohaemagglutinin (PHA). Cell proliferation was measured by MTT assay as previously described by Li et al (24). Absorbance was measured at 570 nm using a Multi-Well Plate Reader (Beckman Coulter Inc., Brea, CA, USA). The proliferation index (PI) of the T cells was calculated using the following equation: PI = mixed lymphocyte reaction/lymphocyte reaction. The experiment was conducted 3 times.

Flow cytometry

The level of surface molecules on DCs was determined by flow cytometry using anti-human antibodies: FITC-CD83, PE-CD86 and APC-HLA-DR (BioLegend, San Diego, CA, USA) as previously described (25). Negative controls were fluorochrome-conjugated isotype-matched irrelevant antibodies (Invitrogen, Carlsbad, CA, USA). Briefly, cells suspended in PBS were incubated with antibodies at room temperature for 30 min in the dark. The cells were then analyzed by a BD FACSCalibur (Becton Dickinson, Franklin Lakes, NJ, USA).

Cytotoxicity assays

Allogeneic T cells were collected as effector cells, and H4M/H2P cells were used as the target cells. Effector cells included the rESO-DC-T group (T cells stimulated with DCs loaded with rESO), the IL-4-DC-T group (T cells stimulated with DCs treated with IL-4) and the T cells group (T cells without stimulation). Effector cells and target cells were incubated at effector/target ratios of 20:1 or 50:1 for 4 h at 37°C in 96-well plates. The activity of T cells against the target tumor cells was measured as previously described in a standard lactate dehydrogenase (LDH) release assay (26). The cytotoxicity of the T cells was calculated as a percentage of specific lysis using the following formula: % specific lysis = (effector/target release − spontaneous release)/(maximal release − spontaneous release) ×100%. Data are presented as the means ± standard deviation.

Immunohistochemistry (IHC)

Formalin-fixed slides from TMAs were deparaffinized by xylene and rehydrated by a series of graded alcohol. Endogenous horseradish peroxidase activity was blocked by treatment with 3% (v/v) H2O2. Antigen retrieval was achieved by heating the samples in a microwave in 10 mM citrate buffer (pH 6.0) for 20 min. Non-specific binding was blocked by incubation with 1% (w/v) BSA in phosphate-buffered saline (PBS) for 1 h at room temperature. The slides were then incubated with 1:200 monoclonal anti-NY-ESO-1 antibody (clone E978; Zymed Laboratories, Inc., South San Francisco, CA, USA) overnight at 4°C. The slides were then incubated with HRP-labeled anti-mouse secondary antibody for 1 h. Immunoreactivity was visualized by diaminobenzidine (DAB). The sections were counterstained with hematoxylin. PBS was used for rinsing between each step. Negative controls were created by omitting the primary antibody. NY-ESO-1 expression was scored by 2 independent observers. The level of NY-ESO-1 expression was described semi-quantitatively using a 4-grade scoring system: -, no staining or focal staining (<5% total); +, 5-<25%; ++, 25–50%; and +++, >50%.

Reverse transcriptase-polymerase chain reaction (RT-PCR)

Total RNA was extracted from the 54 frozen samples and HCC cell lines using TRIzol reagent (Gibco-BRL, Gaithersburg, MD, USA) according to the manufacturer’s instructions. The reverse transcription reaction was performed using the First Strand cDNA Synthesis kit (MBI Fermentas, Vilnius, Lithuania) according to the manufacturer’s instructions. Amplification was carried out using the following primers: ESO-1F (exon 1), 5′-cgcctgcttgagttctacctc-3′; and ESO-1R (exon 3), 5′-agggaaagctgctggagacag-3′. The reaction was conducted under the following conditions: 5 min at 95°C, followed by 30 sec at 94°C, 1 min at 60°C and 45 sec at 72°C for 35 cycles, with a 10 min elongation step at 72°C. β-actin was used as the positive control. The expected PCR product sizes of NY-ESO-1 and β-actin were 219 and 120 bp respectively. Bands were visualized by ethidium bromide staining after separation on a 1.5% agarose gel. The assay was carried out at least 2 times.

Statistical analysis

Statistical analyses were carried out using SPSS version 13.0 software (SPSS, Chicago, IL, USA). Fisher’s exact test or the χ2 test were used to analyze categorical data. Variance analysis was used to determine the statistical significance of the differences between 2 groups. A p-value <0.05 was considered to indicate a statistically significant difference.

Results

DC induction and identification

We obtained 2±0.31×107 mononuclear cells from 50 ml PBMCs following Ficoll separation. The cells were then treated as described in Materials and methods. After 5 days of incubation with GM-CSF and IL-4, 2.2±0.49×106 dendritic-like cells were obtained based on morphology. Flow cytometry analysis revealed that 38.90±2.43% of the imDCs were positive for HLA-DR (Fig. 1). In addition, 3.67±0.49% and 17.23±1.24% of the imDCs were positive for CD83 and CD86, respectively. After rESO or IL-4 induction for 24 h and the addition of TNF-α for 48 h, the rESO-DCs or IL-4-DCs showed a typical branch-like appearance. These cells exhibited a significantly higher expression of HLA-DR, CD83 and CD86 (p<0.05) (Fig. 1). Compared with the imDCs, positivity for HLA-DR, CD83 and CD86 increased by 2- to 21-fold in the rESO-DCs. Similarly, the percentage of HLA-DR, CD83 and CD86 positivity increased by 2- to 17-fold in the IL-4-DCs compared with the imDCs.

To determine the potential of DCs to stimulate T cell proliferation, we performed a mixed T lymphocyte reaction by MTT assay. The PI of the T cells mixed with rESO-DCs was higher than that of those mixed with IL-4-DCs and the imDCs (3.80±0.66 vs. 2.99±0.26 and 1.44±0.36). A statistically significant difference was observed when comparing the rESO-DCs with the imDCs (p<0.05) (Fig. 2). These data illustrated that DCs pulsed with rESO protein were more effective in stimulating T lymphocyte proliferation compared with the control cells.

Cytotoxicity of NY-ESO-1-specific T lymphocytes

The cytotoxicity of T cells against 2 HCC cell lines was determined by LDH release assay. Allogeneic T cells were collected as the effector cells, and H4M and H2P cells were used as the target cells. H4M cells were NY-ESO-1-positive and H2P cells were NY-ESO-1-negative, as shown by RT-PCR (Fig. 3). T cells and HCC cells were incubated at a ratio of 50:1 or 20:1. When the H4M cells were used as the target cells, the specific NY-ESO-1 T cells lysed the cancer cells effectively in a dose-dependent manner (62.13±5.89% for ratio 50:1 and 49.23±3.78% for ratio 20:1), significantly higher than that of the IL-4-DC-T and T cells group (p<0.01) (Fig. 4). By contrast, the specific lysis among the rESO-DC-T, IL-4-DC-T and T cells did not differ significantly in the H2P cells, which did not express NY-ESO-1. These results demonstrate that T cells stimulated with DCs pulsed with rESO exert significant antigen-specific lysis on HCC cells which express NY-ESO-1.

NY-ESO-1 expression in HCC patients

IHC analyses of NY-ESO-1 protein indicated that a total of 30 out of the 190 HCC specimens expressed NY-ESO-1 (15.8%). Among these, 8 (4.2%) were graded as +++, 12 (6.3%) as ++, and 10 (5.3%) as + (Fig. 5A–C). NY-ESO-1 was located predominantly in the cytoplasm, although nuclear staining was observed in a few cells (Fig. 5D). No staining was observed in the tissue adjacent to HCC (Fig. 5E). We also determined the expression profile of NY-ESO-1 mRNA in 54 tumors by RT-PCR. A detectable NY-ESO-1 transcript was observed in 10 tumors (18.5%). Representative results are shown in Fig. 3.

Correlation between NY-ESO-1 expression and clinical parameters

Table II summarizes the correlation between NY-ESO-1 expression and the clinicopathological characteristics of the tumor samples. In the HCC patients with portal vein tumor thrombosis, the frequency of NY-ESO-1 positivity was 24.6% (16/65). By contrast, the frequency of NY-ESO-1 positivity in the HCC patients without portal vein tumor thrombosis was 11.2% (14/125), which was significantly lower compared with the HCC patients with portal vein tumor thrombosis (p=0.013). Statistical analysis also revealed that the frequency of the detectable NY-ESO-1 transcript was higher in HCC patients with portal vein tumor thrombosis as compared with the HCC patients without portal vein tumor thrombosis (33.3 vs. 19.4%, p=0.21). Statistical analysis did not reveal a correlation between NY-ESO-1 expression and age, gender, tumor size, histological grade or hepatitis B surface antigen (HBsAg)/AFP status in the HCC samples.

Table II

NY-ESO-1 expression in the HCC patients.

Table II

NY-ESO-1 expression in the HCC patients.

IHCRT-PCR


GroupNPositive(%)NPositive(%)
Gender
 Male1622515.4441022.7
 Female28517.910330.0
Tumor size
 <5 cm52713.513323.1
 >5 cm1382316.7411024.4
Portal vein tumor thrombosis
 Positive651624.6a18633.3
 Negative1251411.236719.4
Edmondson’s classification
 Grades I–II1031413.629620.7
 Grades III–IV871618.425728
HBsAg
 Positive1552415.5441022.7
 Negative35617.110330.0
AFP
 <20 ng/ml58915.515320.0
 ≥20 ng/ml1322115.9391025.6
Total1903015.8541324.1

a p<0.05 compared with HCC patients without portal vein tumor thrombosis.

{ label (or @symbol) needed for fn[@id='tfn3-ijmm-32-06-1366'] } HCC, hepatocellular carcinoma; IHC, immunohistochemistry; AFP, alpha-fetoprotein.

Correlation between NY-ESO-1 mRNA and protein levels in HCC patients

A total of 54 HCC samples were examined for both the transcript and protein levels of NY-ESO-1. A total of 10 cases were positive for NY-ESO-1 protein, as shown by by IHC and 13 tumors expressed NY-ESO-1 mRNA, as shown by RT-PCR. As presented in Table III, 4 cases were positive for NY-ESO-1 mRNA expression, as shown by RT-PCR, but were shown to be negative by IHC. Out of the 10 tumors with positive immunostaining, 1 tumor was shown to be negative by RT-PCR. This case displayed positive IHC staining for NY-ESO-1.

Table III

Expression of NY-ESO-1 in 54 HCC samples detected by IHC and RT-PCR.

Table III

Expression of NY-ESO-1 in 54 HCC samples detected by IHC and RT-PCR.

IHC

RT-PCR+ (n=5)++ (n=2)+++ (n=3)Negative (n=44)Total
Positive423413
Negative1004041
Total5234454

[i] HCC, hepatocellular carcinoma; IHC, immunohistochemistry; RT-PCR, reverse transcriptase-polymerase chain reaction.

Discussion

The survival of HCC patients is poor despite advancements in HCC therapy (3). In recent years, immunotherapy has become a promising strategy for tumor therapy. A variety of immunotherapy regimens have emerged for HCC patients, including HCC lysates (27), tumor cell-DC fusion (28) and cytokines (29). However, the effects of these methods have been limited. The major barrier to antigen-specific immunotherapy in HCC is a lack of well-defined immunogenic tumor antigens. It has been shown that NY-ESO-1 is one of the most immunogenic CT antigens (7). Immune responses against NY-ESO-1 have been induced in a number of tumors, such as melanoma (30), ovarian cancer (31) and lung cancer (12). The NY-ESO-1 vaccine has been investigated in clinical trials of melanoma and ovarian cancer (32). Recently, a NY-ESO-1 vaccine has also been examined in esophageal and prostate cancer patients (33). Patients bearing NY-ESO-1-expressing tumors displayed an effective induction of NY-ESO-1 antibody and CD4/CD8 T cell responses. Although only a few studies have reported the immune responses induced by NY-ESO-1 in HCC patients (34), NY-ESO-1-specific immune responses in HCC are not yet well understood. In our study, our results demonstrated that NY-ESO-1-specific T cell responses were induced, which significantly increased the lysis of NY-ESO-1-expressing HCC cells in vitro. These data provide evidence supporting the use of NY-ESO-1-based immunotherapy and suggest that NY-ESO-1 may be a useful target for the immunotherapy of HCC patients.

For cancer clinical trials targeted against defined antigens, a detailed knowledge of the antigen expression is crucial. Our data indicated that the positive rate of NY-ESO-1 protein was 15.8% and the NY-ESO-1 mRNA expression was 24.1% in the HCC samples, which was higher than that found in the study by Luo et al (35), and is comparable to the results of Wang et al (13). We also illustrated a high concordance between RT-PCR and IHC for NY-ESO-1 expression in the HCC samples. The HCC patients displayed moderate or high levels of NY-ESO-1 and as shown by IHC, they were positive for the NY-ESO-1 transcript. However, we detected positivity for the NY-ESO-1 transcript in 4 HCC samples without a detectable NY-ESO-1 protein expression. A likely explanation for this is that RT-PCR has a higher sensitivity to detect NY-ESO-1 than IHC. In this study, we also observed heterogeneity for NY-ESO-1 expression by IHC in a few cases, and this information could not be achieved by RT-PCR. It may therefore be desirable to use both IHC and RT-PCR to obtain information regarding the expression level and antigen distribution.

NY-ESO-1 has been speculated to be a prognostic marker in gastrointestinal stromal tumors, melanoma and HCC (15,36,37). NY-ESO-1 expression is associated with a worse tumor outcome. The present study also revealed that NY-ESO-1 expression in HCC patients with portal vein tumor thrombosis had a significantly higher intensity and positivity compared with that in HCC patients without portal vein tumor thrombosis, as shown by IHC and RT-PCR. Thus, it can be hypothesized that the NY-ESO-1 gene may play an important role in tumor invasion and progression. Although the mechanisms involved are unclear, a NY-ESO-1 vaccine may play an important role in the advanced stages of disease, as also found in esophageal carcinoma by Bujas et al (38).

In conclusion, we identified specific T cell responses stimulated with DCs pulsed with NY-ESO-1 in HCC cell lines. We also demonstrate that NY-ESO-1 is heterogeneously expressed in HCC patients, specifically in advanced HCC patients with portal vein tumor thrombosis. This suggests that a DC-based NY-ESO-1 vaccine may prove to be effective for immunotherapy in advanced HCC. Moreover, a combination of demethylation or preparation of multiple CT antigens may increase the efficacy of HCC immunotherapy.

Acknowledgements

This study was supported by the Natural Science Foundation of Fujian Province (C0610023, 2012J01362). We are grateful to Dr JianMing Wen (Department of Pathology, the First Affiliated Hospital, Sun Yat-sen University) for providing the HCC cell lines in this study. We also thank Kay Ka-Wai Li. (Department of Anatomical and Cellular Pathology, the Chinese University of Hong Kong) for revising this article.

References

1 

El-Serag HB and Rudolph KL: Hepatocellular carcinoma: epidemiology and molecular carcinogenesis. Gastroenterology. 132:2557–2576. 2007. View Article : Google Scholar : PubMed/NCBI

2 

Chen MS, Peng ZW, Xu L, Zhang YJ, Liang HH and Li JQ: Role of radiofrequency ablation in the treatment of hepatocellular carcinoma: experience of a cancer center in China. Oncology. 81(Suppl 1): S100–S104. 2011. View Article : Google Scholar : PubMed/NCBI

3 

Rossi L, Zoratto F, Papa A, et al: Current approach in the treatment of hepatocellular carcinoma. World J Gastrointest Oncol. 2:348–359. 2010. View Article : Google Scholar : PubMed/NCBI

4 

Shimozawa N and Hanazaki K: Longterm prognosis after hepatic resection for small hepatocellular carcinoma. J Am Coll Surg. 198:356–365. 2004. View Article : Google Scholar : PubMed/NCBI

5 

Chang CH, Chau GY, Lui WY, Tsay SH, King KL and Wu CW: Long-term results of hepatic resection for hepatocellular carcinoma originating from the noncirrhotic liver. Arch Surg. 139:320–326. 2004. View Article : Google Scholar : PubMed/NCBI

6 

Greten TF, Manns MP and Korangy F: Immunotherapy of HCC. (Review). Rev Recent Clin Trials. 3:31–39. 2008. View Article : Google Scholar

7 

Caballero OL and Chen YT: Cancer/testis (CT) antigens: potential targets for immunotherapy. Cancer Sci. 100:2014–2021. 2009. View Article : Google Scholar : PubMed/NCBI

8 

Hofmann O, Caballero OL, Stevenson BJ, et al: Genome-wide analysis of cancer/testis gene expression. Proc Natl Acad Sci USA. 105:20422–20427. 2008. View Article : Google Scholar : PubMed/NCBI

9 

Gnjatic S, Nishikawa H, Jungbluth AA, et al: NY-ESO-1: review of an immunogenic tumor antigen. Adv Cancer Res. 95:1–30. 2006. View Article : Google Scholar : PubMed/NCBI

10 

Chen YT, Scanlan MJ, Sahin U, et al: A testicular antigen aberrantly expressed in human cancers detected by autologous antibody screening. Proc Natl Acad Sci USA. 94:1914–1918. 1997. View Article : Google Scholar : PubMed/NCBI

11 

Oba-Shinjo SM, Caballero OL, Jungbluth AA, et al: Cancer-testis (CT) antigen expression in medulloblastoma. Cancer Immun. 8:72008.

12 

Kim SH, Lee S, Lee CH, et al: Expression of cancer-testis antigens MAGE-A3/6 and NY-ESO-1 in non-small-cell lung carcinomas and their relationship with immune cell infiltration. Lung. 187:401–411. 2009. View Article : Google Scholar : PubMed/NCBI

13 

Wang XY, Chen HS, Luo S, Zhang HH, Fei R and Cai J: Comparisons for detecting NY-ESO-1 mRNA expression levels in hepatocellular carcinoma tissues. Oncol Rep. 21:713–719. 2009.PubMed/NCBI

14 

Grigoriadis A, Caballero OL, Hoek KS, et al: CT-X antigen expression in human breast cancer. Proc Natl Acad Sci USA. 106:13493–13498. 2009. View Article : Google Scholar : PubMed/NCBI

15 

Svobodová S, Browning J, MacGregor D, et al: Cancer-testis antigen expression in primary cutaneous melanoma has independent prognostic value comparable to that of Breslow thickness, ulceration and mitotic rate. Eur J Cancer. 47:460–469. 2011.

16 

Jäger E, Karbach J, Gnjatic S, et al: Recombinant vaccinia/fowlpox NY-ESO-1 vaccines induce both humoral and cellular NY-ESO-1-specific immune responses in cancer patients. Proc Natl Acad Sci USA. 103:14453–14458. 2006.PubMed/NCBI

17 

Bender A, Karbach J, Neumann A, et al: LUD 00-009: phase 1 study of intensive course immunization with NY-ESO-1 peptides in HLA-A2 positive patients with NY-ESO-1-expressing cancer. Cancer Immun. 7:162007.PubMed/NCBI

18 

Odunsi K, Qian F, Matsuzaki J, et al: Vaccination with an NY-ESO-1 peptide of HLA class I/II specificities induces integrated humoral and T cell responses in ovarian cancer. Proc Natl Acad Sci USA. 104:12837–12842. 2007. View Article : Google Scholar : PubMed/NCBI

19 

Nicholaou T, Ebert L, Davis ID, et al: Directions in the immune targeting of cancer: lessons learned from the cancer-testis Ag NY-ESO-1. Immunol Cell Biol. 84:303–317. 2006. View Article : Google Scholar : PubMed/NCBI

20 

Kakimi K, Isobe M, Uenaka A, et al: A phase I study of vaccination with NY-ESO-1f peptide mixed with Picibanil OK-432 and Montanide ISA-51 in patients with cancers expressing the NY-ESO-1 antigen. Int J Cancer. 129:2836–2846. 2011. View Article : Google Scholar : PubMed/NCBI

21 

Engell-Noerregaard L, Hansen TH, Andersen MH, Thor Straten P and Svane IM: Review of clinical studies on dendritic cell-based vaccination of patients with malignant melanoma: assessment of correlation between clinical response and vaccine parameters. Cancer Immunol Immunother. 58:1–14. 2009. View Article : Google Scholar

22 

Ladhams A, Schmidt C, Sing G, et al: Treatment of non-resectable hepatocellular carcinoma with autologous tumor-pulsed dendritic cells. J Gastroenterol Hepatol. 17:889–896. 2002. View Article : Google Scholar : PubMed/NCBI

23 

Zhang W, Xiao G, Zhang M, Xie D, Guo A and Wen J: The prokaryotic expression, purification and preliminary application of human NY-ESO-1 gene. Zhongguo Kang Ai Xie Hui. 32:626–629. 2005.(In Chinese).

24 

Li DY, Gu C, Min J, Chu ZH and Ou QJ: Maturation induction of human peripheral blood mononuclear cell-derived dendritic cells. Exp Ther Med. 4:131–134. 2012.PubMed/NCBI

25 

Della Bella S, Nicola S, Riva A, Biasin M, Clerici M and Villa ML: Functional repertoire of dendritic cells generated in granulocyte macrophage-colony stimulating factor and interferon-alpha. J Leukoc Biol. 75:106–116. 2004.

26 

Wang XH, Qin Y, Hu MH and Xie Y: Dendritic cells pulsed with gp96-peptide complexes derived from human hepatocellular carcinoma (HCC) induce specific cytotoxic T lymphocytes. Cancer Immunol Immunother. 54:971–980. 2005. View Article : Google Scholar : PubMed/NCBI

27 

Pan K, Zhao JJ, Wang H, et al: Comparative analysis of cytotoxic T lymphocyte response induced by dendritic cells loaded with hepatocellular carcinoma-derived RNA or cell lysate. Int J Biol Sci. 6:639–648. 2010. View Article : Google Scholar : PubMed/NCBI

28 

Cao DY, Yang JY, Yue SQ, et al: Comparative analysis of DC fused with allogeneic hepatocellular carcinoma cell line HepG2 and autologous tumor cells as potential cancer vaccines against hepatocellular carcinoma. Cell Immunol. 259:13–20. 2009. View Article : Google Scholar

29 

Rinaldi M, Iurescia S, Fioretti D, Ponzetto A and Carloni G: Strategies for successful vaccination against hepatocellular carcinoma. Int J Immunopathol Pharmacol. 22:269–277. 2009.PubMed/NCBI

30 

Gedye C, Quirk J, Browning J, et al: Cancer/testis antigens can be immunological targets in clonogenic CD133+ melanoma cells. Cancer Immunol Immunother. 58:1635–1646. 2009. View Article : Google Scholar : PubMed/NCBI

31 

Milne K, Barnes RO, Girardin A, et al: Tumor-infiltrating T cells correlate with NY-ESO-1-specific autoantibodies in ovarian cancer. PLoS One. 3:e34092008. View Article : Google Scholar : PubMed/NCBI

32 

Odunsi K, Matsuzaki J, Karbach J, et al: Efficacy of vaccination with recombinant vaccinia and fowlpox vectors expressing NY-ESO-1 antigen in ovarian cancer and melanoma patients. Proc Natl Acad Sci USA. 109:5797–5802. 2012. View Article : Google Scholar : PubMed/NCBI

33 

Kawada J, Wada H, Isobe M, et al: Heteroclitic serological response in esophageal and prostate cancer patients after NY-ESO-1 protein vaccination. Int J Cancer. 130:584–592. 2012. View Article : Google Scholar : PubMed/NCBI

34 

Korangy F, Ormandy LA, Bleck JS, et al: Spontaneous tumor-specific humoral and cellular immune responses to NY-ESO-1 in hepatocellular carcinoma. Clin Cancer Res. 10:4332–4341. 2004. View Article : Google Scholar : PubMed/NCBI

35 

Luo G, Huang S, Xie X, et al: Expression of cancer-testis genes in human hepatocellular carcinomas. Cancer Immun. 2:112002.PubMed/NCBI

36 

Perez D, Hauswirth F, Jäger D, et al: Protein expression of cancer testis antigens predicts tumor recurrence and treatment response to imatinib in gastrointestinal stromal tumors. Int J Cancer. 128:2947–2952. 2011. View Article : Google Scholar : PubMed/NCBI

37 

Xu H, Gu N, Liu ZB, et al: NY-ESO-1 expression in hepatocellular carcinoma: A potential new marker for early recurrence after surgery. Oncol Lett. 3:39–44. 2012.PubMed/NCBI

38 

Bujas T, Marusic Z, Peric Balja M, Mijic A, Kruslin B and Tomas D: MAGE-A3/4 and NY-ESO-1 antigens expression in metastatic esophageal squamous cell carcinoma. Eur J Histochem. 55:e72011. View Article : Google Scholar : PubMed/NCBI

Related Articles

Journal Cover

December 2013
Volume 32 Issue 6

Print ISSN: 1107-3756
Online ISSN:1791-244X

Sign up for eToc alerts

Recommend to Library

Copy and paste a formatted citation
x
Spandidos Publications style
Chen Y, Huang A, Gao M, Yan Y and Zhang W: Potential therapeutic value of dendritic cells loaded with NY‑ESO‑1 protein for the immunotherapy of advanced hepatocellular carcinoma . Int J Mol Med 32: 1366-1372, 2013
APA
Chen, Y., Huang, A., Gao, M., Yan, Y., & Zhang, W. (2013). Potential therapeutic value of dendritic cells loaded with NY‑ESO‑1 protein for the immunotherapy of advanced hepatocellular carcinoma . International Journal of Molecular Medicine, 32, 1366-1372. https://doi.org/10.3892/ijmm.2013.1510
MLA
Chen, Y., Huang, A., Gao, M., Yan, Y., Zhang, W."Potential therapeutic value of dendritic cells loaded with NY‑ESO‑1 protein for the immunotherapy of advanced hepatocellular carcinoma ". International Journal of Molecular Medicine 32.6 (2013): 1366-1372.
Chicago
Chen, Y., Huang, A., Gao, M., Yan, Y., Zhang, W."Potential therapeutic value of dendritic cells loaded with NY‑ESO‑1 protein for the immunotherapy of advanced hepatocellular carcinoma ". International Journal of Molecular Medicine 32, no. 6 (2013): 1366-1372. https://doi.org/10.3892/ijmm.2013.1510