Open Access

Programmed death-1 blockade enhances the antitumor effects of peptide vaccine-induced peptide-specific cytotoxic T lymphocytes

  • Authors:
    • Yu Sawada
    • Toshiaki Yoshikawa
    • Manami Shimomura
    • Tatsuaki Iwama
    • Itaru Endo
    • Tetsuya Nakatsura
  • View Affiliations

  • Published online on: October 30, 2014     https://doi.org/10.3892/ijo.2014.2737
  • Pages: 28-36
  • Copyright: © Sawada et al. This is an open access article distributed under the terms of Creative Commons Attribution License [CC BY_NC 3.0].

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


Abstract

Novel treatment modalities are required urgently in patients with hepatocellular carcinoma (HCC). A vaccine that induces cytotoxic T lymphocytes (CTLs) is an ideal strategy for cancer, and glypican-3 (GPC3) is a potential option for HCC. Blocking the programmed death-1 (PD-1)/PD-L1 pathway is a rational strategy to overcome tumor escape and tolerance toward CTLs. In the present study, we investigated whether anti-PD-1 blocking antibodies (αPD-1 Ab) enhanced the number of vaccine-induced peptide-specific CTLs in peripheral blood mononuclear cells (PBMCs) following the administration of GPC3 peptide vaccine to both patients and in a mouse model. The inhibitory receptor PD-1 was highly expressed in ex vivo GPC3-specific CTLs isolated from the PBMCs of vaccinated HCC patients. In vitro, interferon-γ induced PD-L1 expression in liver cancer cell lines. In addition, PD-1 blockade increased the number of GPC3-specific CTLs, which degranulate against liver cancer cell lines. In vivo experiments using tumor-bearing mouse models showed that the combination therapy of peptide vaccine and αPD-1 Ab suppressed tumor growth synergistically. PD-1 blockade increased the number of peptide-specific tumor-infiltrating T cells (TILs) and decreased the expression of inhibitory receptors on TILs. This study demonstrated that PD-1/PD-L1 blockade augmented the antitumor effects of a peptide vaccine by increasing the immune response of vaccine-induced CTLs, and provided a foundation for the clinical development of a combination therapy using a GPC3 peptide vaccine and αPD-1 Ab.

Introduction

Antigen-specific cancer immunotherapy using the induction of tumor-specific reactions without autoimmunity is a potentially attractive option for the treatment of cancer. However, immunotherapy for hepatocellular carcinoma (HCC) is still in the preclinical or early clinical trial phases (I and II) of development (1,2). Glypican-3 (GPC3), a carcinoembryonic antigen, is overexpressed in 72–81% of HCC cases, and is correlated with poor prognosis; therefore, it is an ideal target for HCC (37). Recently, a phase I clinical study of a GPC3-derived peptide vaccine reported its safety and efficacy for the treatment of advanced HCC (8). Although vaccine-induced GPC3-peptide-specific cytotoxic T lymphocytes (CTLs) are often tumor reactive in vitro (9) and correlate with overall survival, no complete response was observed when GPC3 peptide vaccination was used as monotherapy in patients with advanced HCC (8).

Programmed death-1 (PD-1) is expressed on activated T and B cells, and elicits inhibitory signals (10). Its ligand PD-L1 is member of the B7 family, and interacts with PD-1 (11). Several studies have shown that the PD-1/PD-L1 pathway plays a critical role in compromised tumor immunity (12,13). PD-1 antibody blockade exerts antitumor effects in clinical trials (14,15). High expression levels of PD-1 on T cells, both in tumor-infiltrating lymphocytes (TILs) and peripheral blood mononuclear cells (PBMCs), were correlated with poor prognosis in HCC patients after surgical resection (16). In addition, PD-L1 expression in HCC was correlated with tumor aggressiveness and postoperative recurrence (17).

In animal models, PD-1 blockade exerts synergistic effects with various tumor vaccines to enhance tumor antigen-specific T cell responses and suppress tumors in vivo (1820). It was reported that melanoma vaccine-induced CTLs become exhausted, which could be reversed by blocking the inhibitory pathways (21). However, a study evaluating the combination of a cancer vaccine and an anti-PD-1 blocking antibody (αPD-1 Ab) for HCC has not been conducted. Therefore, the aim of this study was to investigate whether αPD-1 Ab would enhance the antitumor effects of a peptide vaccine by analyzing CTLs isolated from the PBMCs of vaccinated patients, as well as from a mouse model.

Materials and methods

Patient samples

Three clinical trials were conducted using GPC3-derived peptide vaccines. A phase I trial (n=33) was performed in patients with advanced or metastatic HCC (8) (University Hospital Medical Information Network Clinical Trials Registry; UMIN-CTR no. 000001395). Subsequently, a phase II trial was performed using a GPC3-derived peptide vaccine as an adjuvant therapy in patients with HCC (UMIN-CTR: 000002614, on-going). Finally, a pilot study of liver biopsies taken before and after GPC3 peptide vaccination is being performed for advanced HCC (UMIN-CTR: 000005093, on-going). These trials were approved by the Ethics Committee of the National Cancer Center, Japan, and conformed to the ethical guidelines of the 1975 Declaration of Helsinki. All patients were enrolled after providing written informed consent. Patients were injected intradermally with HLA-A24-restricted GPC3298–306 (EYILSLEEL) or HLA-A2-restricted GPC3144–152 (FVGEFFTDV) peptide vaccines emulsified with incomplete Freund’s adjuvant (IFA, Montanide ISA-51VG; SEPPIC).

Peripheral blood (30 ml) was obtained at the National Cancer Center Hospital East. PBMCs were isolated using standard Ficoll density gradient centrifugation from buffy coats. The remaining PBMCs were used after immunological monitoring in clinical trials. The immunological analyses were approved by the Ethics Committee of the National Cancer Center, Japan.

Cell lines

The human liver cancer cell lines SK-Hep-1 (GPC3, HLA-A*02:01/A*24:02), SK-Hep-1/GPC3 (GPC3+, HLA-A*02:01/A*24:02), and HepG2 (GPC3+, HLA-A*02:01/A*24:02) were available in our laboratory and were used as the target cells (6,9). SK-Hep-1/GPC3 is an established stable GPC3-expressing cell line that was transfected with the human GPC3 gene, whereas SK-Hep-1/vec is an established counterpart cell line that was transfected with an empty vector. The mouse lymphoma cell line RMA (OVA-, H-2Kb) was provided by Dr Yasuharu Nishimura (Kumamoto University, Japan). Cells were cultured at 37°C in RPMI-1640 or DMEM (Sigma-Aldrich) supplemented with 10% fetal bovine serum (FBS), 100 U/ml penicillin and 100 μg/ml streptomycin in a humidified atmosphere containing 5% CO2.

Synthetic peptides and cytokines

The peptides used in this study were as follows: HLA-A*02:01-restricted GPC3144–152 (FVGEFFTDV) peptide (American Peptide Co.), HLA-A*24: 02-restricted GPC3298–306 (EYILSLEEL) peptide (American Peptide Co.), HLA-A*02:01-restricted human immunodeficiency virus (HIV)77–85 (SLYNTYATL) peptide (ProImmune), and H-2Kb-restricted ovalbumin (OVA)257–264 (SIINFEKL) peptide (AnaSpec). The peptides were dissolved and diluted in 7% NaHCO3 or dimethyl sulfoxide (DMSO). Where appropriate, liver cancer cell cultures were treated with 100 U/ml recombinant interferon (IFN)-γ (PeproTech).

Ex vivo Dextramer staining and flow cytometry

PBMCs were stained using HLA-A*02:01 Dextramer-RPE [GPC3144–152 (FVGEFFTDV), HIV19–27 (TLNAWVKVV) or negative control; Immudex] and HLA-A*24:02 Dextramer-RPE [GPC3298–306 (EYILSLEEL), HIV583–591 (RYLKDQQLL); Immudex] for 15 min at room temperature, followed by anti-CD8-FITC (clone T8, Beckman Coulter), anti-PD-1-APC (clone EH12.2H7, BioLegend), or isotype control-APC (clone MOPC-21, BioLegend) for 20 min at 4°C. Flow cytometry was performed using a FACSCanto II (BD Biosciences).

Blocking antibody

GPC3 peptide-specific CTL clones were established from PBMCs as described previously (9). The CTL clones were cultured in AIM-V medium (Life Technologies) supplemented with 10% human AB serum in the presence of 10 μg/ml anti-PD-1 (clone J116, eBioscience) or 10 μg/ml control (clone MOPC-21, BioXcell) monoclonal antibodies for 2 days.

CD107a assay

GPC3 peptide-specific CTL clones were incubated with SK Hep-1/vec pulsed with GPC3144–152 or HIV19–27 peptide and HepG2 at a 1:1 ratio for 3.5 h at 37°C. CTL clones were stained with anti-CD107a-APC (clone LAMP-1, BD Bioscience) during the incubation period, followed by anti-CD8-FITC (clone LT8, ProImmune) for 20 min at 4°C.

Mice

Female C57BL/6 mice (6–8 weeks old) were purchased from Japan Charles River Laboratories (Yokohama, Japan), and were maintained under specific pathogen-free conditions. The Animal Research Committee of the National Cancer Center, Japan, approved all studies. All animal procedures were performed according to the guidelines for the Animal Research Committee of the National Cancer Center, Japan. Ether was used for mouse euthanasia and anesthesia.

In vivo tumor growth inhibition assays

It was reported previously that intratumoral (i.t.) injection of OVA257–264 peptide (SIINFEKL) effectively inhibited OVA-negative tumor growth and survival in a peptide vaccine model using C57BL/6 mice (22). RMA cells (1×105 cells/100 μl PBS) were implanted on the backs of C57BL/6 mouse on day 0. They were then injected with 50-μg peptide mixed with an equal volume of incomplete Freund’s adjuvant (IFA, Montanide ISA-51VG; SEPPIC) on days 7 and 14. The total volume of injected vaccine solution was 100 μl in all experiments. For in vivo therapeutic experiments, anti-mouse PD-1 (clone 4H2) and control Ab (clone MOPC-21, BioXcell) were provided by Ono Pharmaceutical Co., Ltd. The anti-mouse PD-1 Ab (clone 4H2) used in the present study is a chimeric rat Ab containing the murine IgG1 Fc region (23). Anti-PD-1 or control Abs (200 μg/day) were injected intraperitoneally (i.p.) on days 7 and 14. Tumor volume was monitored twice per week, and was calculated using the following formula: tumor volume (mm3) = a × b × b × 0.5, where a is the longest diameter, b is the shortest diameter, and 0.5 is a constant to calculate the volume of an ellipsoid. Mouse health, behavior and mortality were checked daily. All mice were maintained until they showed signs of morbidity or the length or width of the tumors exceeded 30 mm, at which point they were sacrificed for reasons of animal welfare (22).

IFN-γ enzyme-linked immunospot (ELISPOT) analysis

The BDTM ELISPOT set (BD Biosciences) was used to assess the levels of IFN-γ, as described previously (24). Briefly, CD8-positive splenocytes (5×105) were added to the plate as effector cells. Then, either bone marrow-derived dendritic cells (BM-DCs; 1×105) pulsed with OVA peptide (10 μg/ml; as target cells) or non-pulsed BM-DCs (1×105; as control cells) were added. The plate was then incubated for 37°C, for 20 h in the presence of 5% CO2. Spots were counted automatically using the Eliphoto system (Minerva Tech).

Isolation of mouse tumors and flow cytometry

The mice were sacrificed and the dorsal tumors were dissected, cut into small pieces, and digested with collagenase (1 mg/ml) for 20 min at 37°C. After the intratumoral injection of OVA257–264 peptide, tumor cells were isolated and stained with anti-mouse H-2Kb bound to OVA257–264 peptide (SIINFEKL)-PE (clone 25-D1.16, BioLegend) or isotype control-PE (MOPC-21, BioLegend). To analyze the local accumulation of antigen-specific CTLs in mice, isolated tumor cells including tumor-infiltrating lymphocytes were stained with H-2Kb OVA Tetramer-PE [OVA257–264 (SIINFEKL); MBL] for 30 min at room temperature. They were then incubated with anti-mouse CD8-FITC (clone KT15, MBL), anti-mouse PD-1-PE-Cy7 (clone 29F.1A12, BioLegend), anti-mouse CTLA-4-APC (clone UC10-4B, BioLegend), or anti-mouse LAG-3-PerCP-Cy5.5 (clone RTK2071, BioLegend) for 20 min at 4°C.

Quantitative real-time PCR

The tumors implanted into mice were dissected. Total RNA was isolated from homogenized tumors using RNeasy mini kit (Qiagen) according to the manufacturer’s instructions. First-strand complementary deoxyribonucleic acid (cDNA) was synthesized using a PrimeScript® II first-strand cDNA Synthesis kit (Takara). Quantitative real-time PCR was then performed on an Applied Biosystems 7500 FAST Real-time PCR system using Power SYBR® Green (Applied Biosystems). We assessed the expression of the chemokines CXCL10, CXCL12, and CCL3, and compared them to β-actin. Data ware analyzed using delta-delta CT methods. Primer sequences of the chemokines were as described (25), and were purchased from Sigma Genosys.

Statistical analysis

All statistical analyses were performed using PASW Statistics software, version 18.0 (SPSS Inc.). Statistical significance was defined as a value of P<0.05 based on a two-tailed test.

Results

PD-1 expression ex vivo in GPC3 peptide-specific CTLs after vaccination in patients

To investigate whether vaccine-induced CTLs were affected by the PD-1/PD-L1 pathway, we measured the ex vivo expression of PD-1 on vaccine-induced GPC3-specific CTLs using flow cytometry with the GPC3-Dextramer. We used PBMCs obtained from eight patients during clinical trials of the GPC3 peptide vaccine. After vaccination, the frequency of GPC3-specific CTLs increased and could be detected ex vivo, as shown in the representative case 1 (Fig. 1A). GPC3-Dextramer-positive CD8 lymphocytes had a higher expression of PD-1 compared with GPC3-Dextramer-negative CD8 lymphocytes (Fig. 1B; representative case 1). In all eight patients with detectable GPC3-specific CTLs ex vivo after vaccination, PD-1 expression levels were significantly higher in GPC3-Dextramer-positive CD8 lymphocytes compared with GPC3-Dextramer-negative CD8 lymphocytes (Fig. 1C). Before vaccination, no GPC3-Dextramer-positive CD8 lymphocytes were detected ex vivo; therefore, PD-1 expression was not analyzed.

PD-1 blockade augments the GPC3-specific CTL clones that degranulate against liver cancer cell lines

SK-Hep1/vec, SK-Hep1/GPC3, and HepG2 liver cancer cell lines cultured with IFN-γ exhibited marked induction of PD-L1 on their surface (Fig. 2). This suggests that liver cancer cells are invaded by IFN-γ-producing CTLs via the PD-L1-mediated ligation of PD-1. Previously, several GPC3 peptide-specific CTL clones were established from PBMCs isolated from vaccinated patients. These clones exhibited cytotoxic activity against cancer cells expressing GPC3 endogenously (9,26). Therefore, the CD107a (lysosomal-associated membrane protein-1)-mediated externalization of GPC3 peptide-specific CTL clones was examined upon exposure to liver cancer cell lines. The externalization of CD107a could be a surrogate marker to identify the antigen-specific CTLs that degranulate against tumor cells (27). CTL clones mobilized CD107a in response to SK-Hep1/vec pulsed with GPC3144–152 peptide, SK-Hep-1/GPC3, and HepG2 (GPC3+, HLA-A*02:01+), but not in response to pulsed SK-Hep1/vec with HIV19–27 (Fig. 3). Furthermore, PD-1 blockade enriched the population of GPC3-specific CTLs that degranulated against only GPC3-positive liver cancer cell lines (SK-Hep1/vec pulsed with GPC3144–152 peptide, SK-Hep1/GPC3 and HepG2). These results suggest that blocking the interaction between PD-1 and PD-L1 enhanced the antitumor effect of CTLs in liver tumor cells that evade CTLs via PD-L1 expression.

Combination of a peptide vaccine and αPD-1 Ab suppresses tumor growth in vivo synergistically

Intratumoral injection with OVA257–264 peptide (SIINFEKL) effectively inhibited the growth of OVA-negative tumors in a mouse model treated with a peptide vaccine (22). Therefore, we performed in vivo therapeutic experiments using intratumoral OVA peptide vaccine and αPD-1 Ab in tumor implanted mice. Mice were implanted with RMA tumor cells on day 0, and established tumors (3–6 mm in diameter) were treated with OVA peptide emulsified with IFA (vaccine) or vehicle emulsified with IFA (IFA alone) in combination with αPD-1 Ab or control Ab on day 7. An additional dose of vaccine and αPD-1 Ab was administered on day 14 after tumor inoculation (Fig. 4A). On day 21, one mouse in the untreated group was dead, and all other mice were alive. The tumor volume of mice treated using the combination therapy of vaccine and αPD-1 Ab was significantly less than those treated with the appropriate control (Fig. 4B, n=10). Treatment with vaccine/control Ab or IFA alone/αPD-1 Ab did not inhibit tumor growth compared with IFA alone/control Ab treatment. These data suggest that the combination of peptide vaccine and αPD-1 Ab had a synergistic antitumor effect.

Vaccine and αPD-1 Ab treatment increases the number of peptide-specific CTLs within mouse tumors

The loading of injected peptide onto major histocompatibility complex (MHC) class I molecules in tumor cells in vivo was reported previously using IFN-γ ELISPOT assays (22). In the present study, RMA (OVA-, H-2Kb) tumor cells were inoculated onto the backs of C57/BL6 mice. When the tumor diameter reached 3–6 mm, 50 μg H-2Kb-restricted OVA257–264 peptide was injected into the tumor. After 96 h, the tumors were dissected, cut into small pieces, and digested using collagenase. To investigate whether the injected peptide was loaded onto the MHC class I molecules in the tumor cells in a solid mass, flow cytometry using anti-mouse H-2Kb bound to OVA257–264 peptide was performed. The loading of H-2Kb-restricted OVA257–264 peptide onto MHC class I of tumor cells was detected (Fig. 5A).

To evaluate the immunological response to intratumoral OVA peptide vaccine and αPD-1 Ab, the spleens and tumors of mice treated with the same schedule were analyzed as described previously (Fig. 4A). Peptide-specific immune responses were detected in the spleens of mice treated with intratumoral OVA peptide injection using IFN-γ ELISPOT assays (Fig. 5B). Mice that received the combination of intratumoral OVA peptide injection and αPD-1 Ab exhibited an increased number of OVA peptide-specific CTLs compared with those treated with control Ab on day 14 (n=10).

To obtain direct evidence that the combination of peptide vaccine and αPD-1 Ab led to the local accumulation of antigen-specific CTLs, an OVA tetramer assay was performed in mice. OVA-tetramer-positive CD8 lymphocytes could be detected within a tumor using flow cytometry on day 21. Mice that received the combination of OVA peptide vaccine and αPD-1 Ab had a significantly increased number of OVA peptide-specific CTLs compared with those treated with control Ab (Fig. 5C and D; n=8).

Inhibitory receptors on tumor-infiltrating T lymphocytes and the expression of chemokines

The expression of inhibitory receptors on peptide-specific CTLs at the tumor site was assessed to investigate the mechanism of CTL accumulation in the tumors of mice treated with the combination therapy of peptide vaccine and αPD-1 Ab. RMA-bearing mice were treated with intratumoral OVA peptide injection combined with αPD-1 Ab or control Ab, as described previously (Fig. 4A). The expression of PD-1, CTLA-4, and LAG-3 in OVA tetramer-positive CD8 lymphocytes within the tumor on day 21 was analyzed using flow cytometry. The expression of the inhibitory receptors PD-1 and CTLA-4 was decreased in OVA-tetramer positive CD8 lymphocytes in the αPD-1 Ab group compared with the control Ab group (Fig. 6A). However, αPD-1 Ab treatment did not decrease LAG-3 expression in OVA tetramer-positive CD8 lymphocytes.

The expression of chemokines within the tumor on day 21 was examined using quantitative real-time PCR. The expression of the chemokine CCL3 was elevated in mice treated with the combination of intratumoral OVA peptide injection and αPD-1 Ab (Fig. 6B). The expression of the chemokines CXCL10 and CXCL12 was unchanged.

Discussion

Many tumor antigens have been identified in HCC, and their potential clinical utility for the development of cancer-specific immunotherapy has been investigated (2831). GPC3 is a promising target of antigen-specific immunotherapy because it is overexpressed specifically in human HCC (3,4). In addition, it promotes tumor growth by stimulating canonical Wnt signaling (32) or the Hippo pathway (33). A phase I clinical trial of a GPC3-derived peptide vaccine in patients with advanced HCC showed that it had the potential to improve overall survival, which was associated with vaccine-induced CTLs (8). However, the antitumor effects of the peptide-based tumor vaccine alone were not satisfactory in patients with advanced HCC (8,2931). Several studies identified molecules associated with the tumor escape mechanism, such as PD-1/PD-L1, Fas/ FasL, and Decoy receptor 3, which might explain the poor immunogenicity and limitations of the antitumor effects of cancer vaccines alone in patients with advanced HCC (16,17,34,35). Therefore, the present study examined whether blocking PD-1/PD-L enhanced the antitumor effects of peptide vaccines in HCC.

The inhibitory receptor PD-1, was upregulated in GPC3-specific CTLs of HCC patients vaccinated using GPC3 peptide, consistent with previous reports of melanoma vaccine trials (21,27). CTLs for some tumor antigens might not be detected directly ex vivo. The ex vivo analysis of antigen-specific CTLs from uncultured PBMCs could provide strong and novel immunological evidence in HCC vaccine trials. Fourcade et al reported that the upregulation of PD-1 and Tim-3 on CTLs was correlated with the expansion of melanoma-peptide vaccine-induced NY-ESO-1-specific CTLs (21). Further studies are necessary to understand the potential clinical efficacy of vaccine-induced CTLs.

In this experimental model, IFN-γ induced PD-L1 expression in liver cancer cell lines. It was also demonstrated that blocking PD-1 increased the number of GPC3-specific CTL clones that degranulate against these liver cancer cell lines in vitro. These results suggest that blocking the interaction between PD-1 and PD-L1 enhanced the antitumor effects of CTL in liver cancer cells that evaded CTLs by expressing PD-L1. In contrast, Xu et al reported that αPD-L1 or αCTLA-4 Abs did not enhance cytokine secretion and the proliferation of peripheral GPC3-specific CD8+ T-cell from HCC patients significantly (36). Differences in the effects of blocking PD-1 and PD-L1 might account for the differences between spontaneous GPC3-specific CTLs and vaccine-induced CTLs.

The combination of a peptide vaccine with αPD-1 Ab enhanced tumor suppression and antigen-specific T cell infiltration into the tumors of mouse models. The exact mechanisms by which CTLs accumulate into tumors by blocking PD-1 are unclear. A previous study in a mouse model of adoptive cell transfer demonstrated that blocking PD-1 increased the production of CXCL10 by bone marrow-derived myeloid cells, which enhanced the recruitment of CTLs in the tumor (25). We hypothesize that the αPD-1 Ab affected chemokine expression, which resulted in recruitment of vaccine-induced CTLs to the tumor. In the present study, the experimental model did not show a change in the expression of CXCL10. However, the expression of CCL3 was elevated by the combination treatment with vaccine and αPD-1 Ab. Furthermore, blocking PD-1 decreased the expression of inhibitory receptors in peptide-specific CTLs at the tumor site. Recently, mouse models revealed that peptide/IFA vaccination increased the antigen-driven expression of the inhibitory receptors PD-1, LAG-3, CTLA-4, and Tim-3 in CTLs, suggesting partial exhaustion (37). PD-1 blockade might be a rational strategy that could be used to rescue CTLs in a state of exhaustion. Interestingly, αPD-1 Ab therapy did not decrease LAG-3 expression in TILs; however, CTLA-4 expression was decreased, suggesting the partial rescue of CTL from exhaustion. A previous study reported that dual treatment with αLAG-3 and αPD-1 Ab was effective in mice with established tumors (38) as well as during the in vitro expansion of human NY-ESO-1-specific CTLs (39). Furthermore, Sierro et al reported that blocking both PD-1 and PD-L1 might further enhance the antitumor effects of tumor vaccines in mouse models (40).

Based on the results of this clinical trial, the GPC3 peptide vaccine has fewer side effects due to its antigen specificity (8). Enhancing GPC3 peptide vaccine therapy is considered to be promising in terms of sustained tumor control in HCC patients. These data suggest that use of αPD-1 Ab could enhance the antitumor effects of a peptide vaccine, and provide the foundation for the clinical development of a combination therapy.

Acknowledgements

We thank Kayoko Shoda for technical assistance. We also thank Dr Shigehisa Kitano (National Cancer Center), Masashi Minami, Takao Yoshida and Hirotsugu Takano (Ono Pharmaceutical Co.) for scientific advice. Y.S. would like to thank the Foundation for Promotion of Cancer Research (Japan) for the Third-Term Comprehensive Control Research for Cancer for the award of a research resident fellowship. This study was supported in part by the National Cancer Center Research and Development Fund (25-A-7), as well as Research for Promotion of Cancer Control Programmes, Research on Applying Health Technology, and Third Term Comprehensive Control Research for Cancer from the Ministry of Health, Labor and Welfare, Japan and a research funding from Ono Pharmaceutical Co., Ltd. T.N. is a scientific advisor for Ono Pharmaceutical Co., Ltd.

Abbreviations:

HCC

hepatocellular carcinoma

CTL

cytotoxic T lymphocyte

GPC3

glypican-3

PD-1

programmed death-1

PBMC

peripheral blood mononuclear cell

HLA

human leukocyte antigen

IFN-γ

interferon-γ

MHC

major histocompatibility complex

References

1 

Breous E and Thimme R: Potential of immunotherapy for hepatocellular carcinoma. J Hepatol. 54:830–834. 2011. View Article : Google Scholar

2 

Greten TF, Manns MP and Korangy F: Immunotherapy of hepatocellular carcinoma. J Hepatol. 45:868–878. 2006. View Article : Google Scholar : PubMed/NCBI

3 

Nakatsura T, Yoshitake Y, Senju S, et al: Glypican-3, overexpressed specifically in human hepatocellular carcinoma, is a novel tumor marker. Biochem Biophys Res Commun. 306:16–25. 2003. View Article : Google Scholar : PubMed/NCBI

4 

Capurro M, Wanless IR, Sherman M, et al: Glypican-3: a novel serum and histochemical marker for hepatocellular carcinoma. Gastroenterology. 125:89–97. 2003. View Article : Google Scholar : PubMed/NCBI

5 

Nakatsura T, Komori H, Kubo T, et al: Mouse homologue of a novel human oncofetal antigen, glypican-3, evokes T-cell-mediated tumor rejection without autoimmune reactions in mice. Clin Cancer Res. 10:8630–8640. 2004. View Article : Google Scholar

6 

Komori H, Nakatsura T, Senju S, et al: Identification of HLA-A2- or HLA-A24-restricted CTL epitopes possibly useful for glypican-3-specific immunotherapy of hepatocellular carcinoma. Clin Cancer Res. 12:2689–2697. 2006. View Article : Google Scholar : PubMed/NCBI

7 

Shirakawa H, Suzuki H, Shimomura M, et al: Glypican-3 expression is correlated with poor prognosis in hepatocellular carcinoma. Cancer Sci. 100:1403–1407. 2009. View Article : Google Scholar : PubMed/NCBI

8 

Sawada Y, Yoshikawa T, Nobuoka D, et al: Phase I trial of a glypican-3-derived peptide vaccine for advanced hepatocellular carcinoma: immunologic evidence and potential for improving overall survival. Clin Cancer Res. 18:3686–3696. 2012. View Article : Google Scholar

9 

Yoshikawa T, Nakatsugawa M, Suzuki S, et al: HLA-A2-restricted glypican-3 peptide-specific CTL clones induced by peptide vaccine show high avidity and antigen-specific killing activity against tumor cells. Cancer Sci. 102:918–925. 2011. View Article : Google Scholar

10 

Agata Y, Kawasaki A, Nishimura H, et al: Expression of the PD-1 antigen on the surface of stimulated mouse T and B lymphocytes. Int Immunol. 8:765–772. 1996. View Article : Google Scholar : PubMed/NCBI

11 

Freeman GJ, Long AJ, Iwai Y, et al: Engagement of the PD-1 immunoinhibitory receptor by a novel B7 family member leads to negative regulation of lymphocyte activation. J Exp Med. 192:1027–1034. 2000. View Article : Google Scholar : PubMed/NCBI

12 

Iwai Y, Ishida M, Tanaka Y, Okazaki T, Honjo T and Minato N: Invovement of PD-L1 on tumor cells in the escape from host immune system and tumor immunotherapy by PD-L1 blockade. Proc Natl Acad Sci USA. 99:12294–12297. 2002.PubMed/NCBI

13 

Iwai Y, Terawaki S and Honjo T: PD-1 blockade inhibits hematogenous spread of poorly immunogenic tumor cells by enhanced recruitment of effector T cells. Int Immunol. 17:133–144. 2005. View Article : Google Scholar : PubMed/NCBI

14 

Topalian SL, Hodi FS, Brahmer JR, et al: Safety, activity, and immune correlates of anti-PD-1 antibody in cancer. N Engl J Med. 366:2443–2454. 2012. View Article : Google Scholar : PubMed/NCBI

15 

Wolchok JD, Kluger H, Callahan MK, et al: Nivolumab plus ipilimumab in advanced melanoma. N Engl J Med. 369:122–133. 2013. View Article : Google Scholar : PubMed/NCBI

16 

Shi F, Shi M, Zeng Z, et al: PD-1 and PD-L1 upregulation promotes CD8(+) T-cell apoptosis and postoperative recurrence in hepatocellular carcinoma patients. Int J Cancer. 128:887–896. 2011. View Article : Google Scholar : PubMed/NCBI

17 

Gao Q, Wang XY, Qiu SJ, et al: Overexpression of PD-L1 significantly associates with tumor aggressiveness and postoperative recurrence in human hepatocellular carcinoma. Clin Cancer Res. 15:971–979. 2009. View Article : Google Scholar : PubMed/NCBI

18 

McGray AJ, Bernard D, Hallett R, et al: Combined vaccination and immunostimulatory antibodies provides durable cure of murine melanoma and induces transcriptional changes associated with positive outcome in human melanoma patients. Oncoimmunology. 1:419–431. 2012. View Article : Google Scholar

19 

Mkrtichyan M, Najjar YG, Raulfs EC, et al: Anti-PD-1 synergizes with cyclophosphamide to induce potent antitumor vaccine effects through novel mechanisms. Eur J Immunol. 41:2977–2986. 2011. View Article : Google Scholar : PubMed/NCBI

20 

Duraiswamy J, Kaluza KM, Freeman GJ and Coukos G: Dual blockade of PD-1 and CTLA-4 combined with tumor vaccine efficacy restorres T cell rejection function in tumors. Cancer Res. 73:3591–3603. 2013. View Article : Google Scholar : PubMed/NCBI

21 

Fourcade J, Sun Z, Pagliano O, et al: PD-1 and Tim-3 regulate the expansion of tumor antigen-specific CD8+ T cells induced by melanoma vaccines. Cancer Res. 74:1045–1055. 2014. View Article : Google Scholar : PubMed/NCBI

22 

Nobuoka D, Yoshikawa T, Takahashi M, et al: Intratumoral peptide injection enhances tumor cell antigenicity recognized by cytotoxic T lymphocytes: a potential option for improvement in antigen-specific cancer immunotherapy. Cancer Immunol Immunother. 62:639–652. 2013. View Article : Google Scholar

23 

Li B, VanRoey M, Wang C, Chen TH, Korman A and Jooss K: Anti-programmed death-1 synergizes with granulocyte macrophage colony-stimulating factor - secreting tumor cell immunotherapy providing therapeutic benefit to mice with established tumors. Clin Cancer Res. 15:1623–1634. 2009. View Article : Google Scholar

24 

Iwama T, Horie K, Yoshikawa T, et al: Identification of an H2-Kb or H2-Db restricted and glypican-3-derived cytotoxic T-lymphocyte epitope peptide. Int J Oncol. 42:831–838. 2013.

25 

Peng W, Liu C, Xu C, et al: PD-1 blockade enhances T-cell migration to tumors by elevating IFN-γ inducible chemokines. Cancer Res. 72:5209–5218. 2012. View Article : Google Scholar : PubMed/NCBI

26 

Tada Y, Yoshikawa T, Shimomura M, et al: Analysis of cytotoxic T lymphocytes from a patient with hepatocellular carcinoma who showed a clinical response to vaccination with a glypican-3-derived peptide. Int J Oncol. 43:1019–1026. 2013.

27 

Wong RM, Scotland RR, Lau RL, et al: Programmed death-1 blockade enhances expansion and functional capacity of human melanoma antigen-specific CTLs. Int Immunol. 19:1223–1234. 2007. View Article : Google Scholar : PubMed/NCBI

28 

Mizukoshi E, Nakamoto Y, Arai K, et al: Comparative analysis of various tumor-associated antigen-specific T-cell responses in patients with hepatocellular carcinoma. Hepatology. 53:1206–1216. 2011. View Article : Google Scholar : PubMed/NCBI

29 

Butterfield LH, Ribas A, Meng WS, et al: T-cell responses to HLA-A*0201 immunodominant peptides derived from alpha-fetoprotein in patients with hepatocellular cancer. Clin Cancer Res. 9:5902–5908. 2003.PubMed/NCBI

30 

Butterfield LH, Ribas A, Dissette VB, et al: A phase I/II trial testing immunization of hepatocellular carcinoma patients with dendritic cells pulsed with four alpha-fetoprotein peptides. Clin Cancer Res. 12:2817–2825. 2006. View Article : Google Scholar

31 

Greten TF, Forner A, Korangy F, et al: A phase II open trial evaluating safety and efficacy of a telomerase peptide vaccination in patients with advanced hepatocellular carcinoma. BMC Cancer. 10:2092010. View Article : Google Scholar : PubMed/NCBI

32 

Capurro MI, Xiang YY, Lobe C and Filmus J: Glypican-3 promotes the growth of hepatocellular carcinoma by stimulating canonical Wnt signaling. Cancer Res. 65:6245–6254. 2005. View Article : Google Scholar : PubMed/NCBI

33 

Feng M, Gao W, Wang R, et al: Therapeutically targeting glypican-3 via a conformation-specific single-domain antibody in hepatocellular carcinoma. Proc Natl Acad Sci USA. 110:E1083–E1091. 2013. View Article : Google Scholar : PubMed/NCBI

34 

Nagao M, Nakajima Y, Hisanaga M, et al: The alteration of Fas receptor and ligand system in hepatocellular carcinomas: how do hepatoma cells escape from the host immune surveillance in vivo? Hepatology. 30:413–421. 1999. View Article : Google Scholar : PubMed/NCBI

35 

Chen C, Zhang C, Zhuang G, et al: Decoy receptor 3 overexpression and immunologic tolerance in hepatocellular carcinoma (HCC) development. Cancer Invest. 26:965–974. 2008. View Article : Google Scholar : PubMed/NCBI

36 

Xu Y, Li H, Gao RL, Adeyemo O, Itkin M and Kaplan DE: Expansion of interferon-gamma-producing multifunctional CD4+ T-cells and dysfunctional CD8+ T-cells by glypican-3 peptide library in hepatocellular carcinoma patients. Clin Immunol. 139:302–313. 2011. View Article : Google Scholar : PubMed/NCBI

37 

Hailemichael Y, Dai Z, Jaffarzad N, et al: Persistent antigen at vaccination sites induces tumor-specific CD8(+) T cell sequestration, dysfunction and deletion. Nat Med. 19:465–472. 2013. View Article : Google Scholar : PubMed/NCBI

38 

Woo SR, Turnis ME, Goldberg MV, et al: Immune inhibitory molecules LAG-3 and PD-1 synergistically regulate T-cell function to promote tumoral immune escape. Cancer Res. 72:917–927. 2012. View Article : Google Scholar : PubMed/NCBI

39 

Matsuzaki J, Gnjatic S, Mhawech-Fauceglia P, et al: Tumor-infiltrating NY-ESO-1-specific CD8+ T cells are negatively regulated by LAG-3 and PD-1 in human ovarian cancer. Proc Natl Acad Sci USA. 107:7875–7880. 2010. View Article : Google Scholar : PubMed/NCBI

40 

Sierro SR, Donda A, Perret R, et al: Combination of lentivector immunization and low-dose chemotherapy or PD-1/PD-L1 blocking primes self-reactive T cells and induces antitumor immunity. Eur J Immunol. 41:2217–2228. 2011. View Article : Google Scholar : PubMed/NCBI

Related Articles

Journal Cover

January-2015
Volume 46 Issue 1

Print ISSN: 1019-6439
Online ISSN:1791-2423

Sign up for eToc alerts

Recommend to Library

Copy and paste a formatted citation
x
Spandidos Publications style
Sawada Y, Yoshikawa T, Shimomura M, Iwama T, Endo I and Nakatsura T: Programmed death-1 blockade enhances the antitumor effects of peptide vaccine-induced peptide-specific cytotoxic T lymphocytes. Int J Oncol 46: 28-36, 2015
APA
Sawada, Y., Yoshikawa, T., Shimomura, M., Iwama, T., Endo, I., & Nakatsura, T. (2015). Programmed death-1 blockade enhances the antitumor effects of peptide vaccine-induced peptide-specific cytotoxic T lymphocytes. International Journal of Oncology, 46, 28-36. https://doi.org/10.3892/ijo.2014.2737
MLA
Sawada, Y., Yoshikawa, T., Shimomura, M., Iwama, T., Endo, I., Nakatsura, T."Programmed death-1 blockade enhances the antitumor effects of peptide vaccine-induced peptide-specific cytotoxic T lymphocytes". International Journal of Oncology 46.1 (2015): 28-36.
Chicago
Sawada, Y., Yoshikawa, T., Shimomura, M., Iwama, T., Endo, I., Nakatsura, T."Programmed death-1 blockade enhances the antitumor effects of peptide vaccine-induced peptide-specific cytotoxic T lymphocytes". International Journal of Oncology 46, no. 1 (2015): 28-36. https://doi.org/10.3892/ijo.2014.2737