Mucin 1 and poly I:C activates dendritic cells and effectively eradicates pituitary tumors as a prophylactic vaccine

  • Authors:
    • Dehua Sui
    • Lixin Ma
    • Meng Li
    • Wei Shao
    • Hongpeng Du
    • Ke Li
    • Zhenzhu Li
    • Zefu Li
  • View Affiliations

  • Published online on: March 2, 2016     https://doi.org/10.3892/mmr.2016.4964
  • Pages: 3675-3683
Metrics: Total Views: 0 (Spandidos Publications: | PMC Statistics: )
Total PDF Downloads: 0 (Spandidos Publications: | PMC Statistics: )


Abstract

Pituitary tumors are the most common type of cancer within the central nervous system. In the present study, the expression levels of mucin 1 (Muc1) were examined in invasive and non‑invasive pituitary tumor samples, and the results of immunohistochemical staining and Western blot analysis demonstrated marked positive expression of Muc1. In addition, Muc1 + polyinosinic:polycytidylic acid (poly I:C) was found to stimulate the expression levels of the surface molecules cluster of differentiation (CD)40, CD83 and CD80, and HLA‑DRm and decreased the expression of CD14 in the dendritic cells, determined using fluorescence‑activated cell sorting. The secretions of interleukin (IL)‑6, tumor necrosis factor‑α and IL‑1β cytokines were also significantly induced, in a dose‑dependent manner. In in vivo experiments, a higher percentage of CD3+CD4+ T lymphocytes was detected, and the levels of interferon‑γ and IL‑2 in the splenocytes were also upregulated. Furthermore, the combination treatment of Muc1 with poly I:C increased anti‑Muc1 IgM and anti‑Muc1 IgG titers, and altered the balance of IgG2a and IgG1, all of which increased the T helper (Th)1 polarized immune response. Thus, the tumor antigen, Muc1, with poly I:C may produce potent protective effects, which polarize immune responses towards Th1, and elicit antitumor immunity to inhibit the progression of pituitary tumors.

Introduction

Pituitary tumors account for between 10 and 25% of all intracranial neoplasms (1). They represent the third most common type of cancer within the central nervous system, and is only less frequent than glioma and meningioma (1,2). Pituitary tumors are also one of the most common tumors of the endocrine system (3). With the continuous development of diagnostic imaging and laboratory diagnosis, the incidence of pituitary tumors has gradually increased (4). Clinically, the most common classification methods for pituitary tumors are based on their endocrine function and immunohistochemistry staining analysis. Pituitary adenomas are classified based upon anatomical, histological and functional criteria (5), including lactotrophic adenomas (prolactinomas), somatotrophic adenomas, corticotrophicadenomas, gonadotrophicadenomas, thyrotrophic adenomas (rare) and null cell adenomas (6,7). Lactotrophic adenomas, secreting prolactin, account for 30%, somatotrophic adenomas account for 15% and non-secretive null cell adenomas account for 25% of pituitary adenomas (8,9).

Although several techniques and drugs have been investigated and used in the treatment of pituitary cancer, there has been no significant improvement in the local control of carcinoma development and progression, and there have been no significant improvements in survival rates or in quality of life of patients with pituitary cancer (10,11). Following its rapid development in cancer therapy, immunotherapy has generally become one of the important treatment strategies following surgery, radiation therapy and chemotherapy (12,13). Provenge is a dendritic cell (DC)-based therapeutic cancer vaccine, which was approved by the Food and Drug Administration in April 2010 (14,15), until which DCs were first approved for the treatment of advanced prostate cancer. Currently, DC immunotherapy is broadly used in clinical therapy, and is used in the treatment of breast cancer, lung cancer, gastrointestinal cancer, prostate cancer, kidney cancer and malignant melanoma (1620). DC immunotherapy has a broad range of applications, is safe and has no significant adverse reactions (21,22).

Mucins are a family of high molecular weight, heavily glycosylated proteins (23). To date, over 20 mucin genes have been identified and cloned, including membrane-bound Muc1, Muc3, Muc4 and Muc12, and secreted Muc2, Muc5, Muc6 and Muc9 (24,25). Muc1 is an epithelial membrane associated glycoprotein (Episialin), and it is the first mucin to be characterized and cloned in detail (26). An increasing number of mucins are being used as important molecular markers. In the present study, polyinosinic:polycytidylic acid (poly I:C) was used as an immunostimulatory agent or immune adjuvant. The present study also aimed to investigate whether Muc1 offers potential as a candidate vaccine antigen to prevent or control the progression of pituitary tumors in vitro and in vivo.

Materials and methods

Cell line and reagents

The AtT20 cell line was obtained from American Type Culture Collection (cat. no. CRL1795), and the cells were cultured in Dulbecco's modified Eagle's medium (DMEM; Invitrogen; Thermo Fisher Scientific, Inc., Waltham, MA, USA) supplemented with 10% fetal bovine serum (FBS; Thermo Trace, Ltd., Melbourne, Australia) at 37° C in an atmosphere of 95% air and 5% CO2. Human recombinant Muc1 protein was obtained from Abnova Corporation (Tapei City, Taiwan). Mouse anti-human MUC1 (sc-59795) and anti-human β-actin (sc-69879) monoclonal antibodies (mAbs), and horseradish peroxidase (HRP)-conjugated goat anti-mouse secondary antibody (sc-2031) were obtained from Santa Cruz Biotechnology, Inc (Santa Cruz, CA, USA).

Patients

A total of 42 patients with invasive pituitary adenoma, and 44 patients with non-invasive pituitary adenoma, at the Affiliated Hospital of Binzhou Medical College (Binzhou, China) were enrolled in the present study. Pituitary adenoma specimens were collected during surgery in order to detect the expression levels of Muc1 by immunohistochemistry and western blotting. The samples were maintained at −80° C until further analysis. The present study was approved by the ethics committee of Affiliated Hospital of Binzhou Medical College. Written informed consent was obtained from all patients.

Immunohistochemical analysis

Immunohistochemical staining was performed in order to detect the expression of Muc1 in the pituitary adenoma specimens. Paraffin-embedded sections were dewaxed, rehydrated, blocked and incubated overnight with mouse anti-human MUC1 (1:1,000) and anti-human β-actin (1:1,000) mAbs. Subsequently, the specimens were washed three times and incubated with HRP-conjugated goat anti-mouse secondary antibody (1:1,000) for 1 h. The stained slides were dehydrated, made transparent, mounted in Permount and visualized using a Nikon Eclipse Ti-E microscope (Nikon Corporation, Tokyo, Japan). The images were captured with a camera attached to the microscope at ×400 magnification.

Western blotting

The invasive and non-invasive pituitary adenoma specimens were lysed using radioimmunoprecipitation assay buffer (Beyotime Institute of Biotechnology, Shanghai, China) consisting of 50 mmol/l Tris, 1% NP-40, 150 mmol/l NaCl, 1.0 mmol/l EDTA, 0.1% sodium dodecyl sulfate (SDS) and 0.25% SDC. Equal quantities of protein (10 µg) were separated by 10% SDS-polyacrylamide gel electrophoresis and blotted onto polyvinylidene difluoride membranes. After blocking the membranes with 5% bovine serum albumin (Beyotime Institute of Biotechnology) in phosphate-buffered saline, the membranes were incubated with mouse anti-MUC1 and anti-β-actin mAbs (1:1,000) at 4° C overnight. Subsequently, the membranes were washed three times with PBS containing 0.05% Tween-20 (PBST), followed by incubation with HRP-conjugated goat anti-mouse secondary antibody (1:1,000) at room temperature for 40 min. The membranes were visualized using the Pierce ECL Western Blotting Substrate (cat. no. 32209; Thermo Fisher Scientific, Inc.), and blot intensities were analyzed using ImageJ software, version 2 (National Institutes of Health, Bethesda, MA, USA).

Phenotype identification of peripheral blood mononuclear cell (PBMC)-derived DCs by fluorescence-activated cell sorting (FACS) analysis

The DCs were generated and cultured from human PBMCs, as described in a previous study (27). The PBMCs were obtained from blood samples derived from healthy donors using Ficoll-Hypaque density gradient centrifugation at 300 x g for 5 min at 37° C. The PBMCs (5×104 cells/well) were plated into 6-well plates. Following 5 days of culture, human immature DCs were stimulated with 50 µg/ml Muc1 and 50 µg/ml poly I:C, or with 50 µg/ml Muc1 alone for 48 h at 37° C. Subsequently, the cells were collected and washed with PBS for phenotypic analysis. The DCs were stained with phycoerythrin (PE)-conjugated mAbs against CD40 (cat. no. 313005), CD80 (cat. no. 305207) and CD83 (cat. no. 305307), and fluorescein isothiocyanate (FITC)-conjugated mAbs against CD14 (cat. no. 325603) and histocompatability locus antigen (HLA)-DR (cat. no. 307603), as well as allophycocyanin anti-human CD86 mAb (cat. no. 305411; all BioLegend, San Diego, CA, USA) for 20 min at 4° C (all 1:1,000).

Determination of DC cytokine production using an enzyme-linked immunosorbent assay (ELISA)

The human immature DCs were adjusted to a density of 5×105 cells/ml and plated into 24-well plates. The cells were treated with Muc1, Muc1 + poly I:C or lipopolysaccharide (LPS) for 48 h, and the culture supernatants were harvested at the end of the experiment at 37° C, and centrifuged at 300 × g for 5 min at 37° C, prior to ELISA assay. The levels of the cytokines tumor necrosis factor (TNF)-α, interleukin (IL)-1β and IL-6 in the supernatants were measured, according to the manufacturer's protocol (Neobioscience Technology, Co., Ltd., Beijing, China).

Mice and immunization

A total of 18 C57BL/6 male mice (6–8 weeks old) were obtained from the Laboratory Animal Center of Peking University Health Science Center (Beijing, China). The mice were housed in specific pathogen-free conditions under a 12-h light/dark cycle at 21–25° C, and with ad libitum access to food and water. The mice were randomly assigned to three groups (6 mice/group), as follows: i) The Muc1 group; ii) the Muc1 + poly I:C group; and iii) the control group. The mice were twice injected subcutaneously on days 1 and 15 with 20 µg Muc1 antigen, 20 µg Muc1 antigen + 50 µg poly I:C or with 100 µl PBS. The mice were sacrificed by cervical dislocation 7 days following the final immunization and the abdominal cavity was opened with scissors in order to obtain the spleens. Splenocyte cell suspensions were prepared using a sterile syringe core and 200 mesh steel filter.

Previous studies have reported that tumor cell lysate (TLA), which is used as a source of tumor-specific antigens, is able to induce the maturation of dendritic cells; although the maturation-inducing effect of TLAs was not particularly strong (11,12). In the present study, the mice were vaccinated with TLA or TLA + poly I:C, and the protein expression levels of interferon (IFN)-γ and IL-2 in the splenocytes of the mice were investigated using the Mouse IFN-γ ELISA (cat. no. EM0560) and the Mouse IL-2 ELISA kit (cat. no. EM0290), respectively (both Beijing Solarbio Science & Technology Co., Ltd., Beijing, China). The TLA was obtained from the AtT20 cells. Briefly, cells were washed once with PBS, harvested and then resuspended in PBS at 1×107 cells/ml, prior to 5 cycles of freezing using methanol with dry ice and thawing at 56° C. Subsequently, the cells were harvested, and washed in PBS, prior to centrifugation at 1,700 × g for 5 min. The TLAs were contained in the supernatant, which were separated and stored at −80° C prior to vaccination of the mice.

T-cell subtype analysis by FACS analysis

The T-cell subtype was determined using FACS analysis. At 10 days following the second immunization (day 15), blood samples were taken from the venous plexus of the mouse eyes and 50 µl heparin-treated orbital blood was lysed with red blood cell lysis buffer (eBioscience, San Diego, CA, USA). Lymphocytes were stained with 100 µl PBS containing 1% BSA and 0.1% NaN3, together with 5 µl FITC-conjugated anti-CD3 mAb (cat. no. 16-0037-81), followed by simultaneous staining with 5 µl of PE-labeled anti-CD4 (cat. no. 12-0048-41) or anti-CD8 (cat. no. 12-0809-41) mAbs (all eBioscience). The cells were then incubated for 20 min at 4° C, and flow cytometry was performed using the BD FACSCalibur flow cytometer (BD Biosciences, Franklin Lakes, NJ, USA).

Measurement of Muc1-specific antibodies

Muc1-specific antibodies in the mouse serum were measured using an ELISA (28). Firstly, microtiter plates (Nalge Nunc International, Penfield, NY, USA) were coated with Muc1 (10 µg/ml) in 50 mM carbonate buffer (pH 9.6) overnight at 4° C. Following washing with PBST, the wells were blocked with 100 µl 5% dried milk in PBST for 4 h at 37° C. To measure IgM, IgG, IgG2a and IgG1, the plates containing with serum samples were incubated with HRP-conjugated rabbit anti-mouse IgG for 30 min at 37° C. Following incubation with substrate buffer, the enzyme reaction was terminated after 5 min by adding 50 µl 2 M H2SO4. The absorbance at 450 nm was then measured using the Multiskan MK3 microplate reader (Thermo Fisher Scientific, Inc.). All washes were performed using PBS-T. Serum samples and HRP-conjugated rabbit anti-mouse IgG (sc-358914; 1:2,000; Santa Cruz Biotechnology, Inc.) were diluted in PBS-T containing 3% dried milk.

Cytotoxicity assay

Lactate dehydrogenase (LDH) release assays were performed according to the manufacturer's protocol (KeyGen Biotech Co., Ltd., Nanjing, China). The C57BL/6 mice were immunized, as described above and, 10 days following the final immunization, splenocytes were prepared as effector cells, and AtT20 cells were used as target cells. Cytolytic T lymphocyte activity was measured by a LDH release assay in 96-well round-bottom plates. Target cells (2×104 cells per well) in a volume of 100 µl were incubated with 100 µl effector cells at 5/1, 10/1 or 20/1 effector:target (E:T) ratios for 24 h at 37° C in phenol red-free DMEM containing 2% FBS. Following centrifugation at 300 × g for 5 min at 37° C, the supernatants (50 µl per well) were transferred into 96-well flat-bottom plates for measurement of absorbance at 490 nm using the Multiskan MK3 microplate reader.

Statistical analysis

Data were analyzed using SPSS software, version 19.0 (IBM SPSS, Armonk, NY, USA). Data are presented as the mean ± standard deviation. Statistical significance in the different treatment groups was compared using a log-rank test. P<0.05 was considered to indicate a statistically significant difference.

Results

Positive Muc1 staining is observed in invasive pituitary adenoma

In the present study, 42 registered cases of invasive pituitary adenoma, and 44 samples of non-invasive pituitary adenoma were included from the hospital database. The expression of Muc1 was detected using immunohistochemistry in the invasive and non-invasive pituitary adenomas, respectively. As shown in Fig. 1A, marked positive expression of Muc1 was observed, with dark brown or brown particles, which were predominantly located in the cytoplasm and membrane. By contrast, negative or weak staining of Muc1 was observed in the non-invasive pituitary adenomas. The positive rates of Muc1 expression in the invasive and non-invasive pituitary adenomas were 90.4% (32/42) and 20.5% (9/44), respectively (Table I). There was a significant difference between the two groups (P<0.01). The expression levels of Muc1 were also determined in samples of non-invasive pituitary tissues and malignant pituitary adenomas by western blotting, and the results were consistent with those obtained by the immunohistochemical staining (Fig. 1B).

Table I

Comparison of the expression levels of mucin 1 in pituitary adenoma tissues.

Table I

Comparison of the expression levels of mucin 1 in pituitary adenoma tissues.

Type of tissueNegative expressionWeak positive expressionStrong positive expressionPositivity (%)
Invasive pituitary adenoma463290.4
Non-invasive pituitary adenoma359020.5
Production of cytokines in human DCs in the Muc1 + poly I:C group and poly I:C group

In the present study, DCs were derived from adherent PBMCs cultured in medium supplemented with human granulocyte-macrophage colony-stimulating factor and IL-4, and the DCs are shown in Fig. 2A. As Muc1 was expressed at high levels in the malignant pituitary adenomas, Muc1 was used as the tumor antigen, and poly I:C was selected as an immune stimulator agent. Cytokine secretion was examined following stimulation of the immature PBMC-DCs with Muc1 or different concentrations of Poly I:C, and the resulting supernatants were harvested after 48 h. As shown in Fig. 2B–D, poly I:C significantly enhanced the secretion of IL-1β, TNF-α and IL-6 in a dose-dependent manner, as determined using ELISA. In this experiment, untreated cells were used as negative controls and cells treated with LPS were used as positive controls.

Phenotypic identification of PBMC-derived DCs

In order to assess the phenotypic variation of the human DCs, the expression of surface markers on the PBMC-derived DCs was examined using FACS analysis. The immature DCs were treated with either 50 µg/ml poly I:C or with 50 µg/ml poly I:C + 50 µg/ml Muc1 for 48 h, following which the expression levels of costimulatory molecules (CD40 and CD80), HLA-DR and the CD83 maturation marker were measured. As shown in Fig. 3A and B, the results demonstrated that poly I:C or Muc1-poly I:C induced the phenotypic maturation of the human DCs, with low expression levels of CD14, moderate expression levels of CD40, CD80 and CD83, and marked expression of HLA-DR.

Determination of CD4+CD3+ T cell subpopulations using FACS analysis

In order to detect the immune response in vivo, C57BL/6 mice were used as an animal model. As is already known, T-cell subsets include T helper cells (CD4+CD3+ T lymphocytes) and cytotoxic T cells (CD8+CD3+ T lymphocytes) (29). As shown in Fig. 4A–D, the mice immunized with Muc1 + poly I:C contained a higher percentage of CD3+CD4+ T lymphocytes in their peripheral blood, compared with the mice injected with the negative control, PBS. However, the percentage of CD8+CD3+ T lymphocytes in the peripheral blood did not change significantly (data not shown).

Mice immunized with Muc1 + poly I:C have increased Muc1-specific serum antibody titers following vaccination

The present study also measured antigen-specific antibody titers in the immunized mice. Muc1 antigen was injected subcutaneously into mice, together with poly I:C, with Muc1 antigen only or with LPS and 10 days following the final immunization, the levels of IgM, total IgG, IgG2a and IgG1 of the anti-HBs antibodies were examined. As shown in Fig. 5, the mice immunized with Muc1 + poly I:C had a higher geometric mean titer of Muc1 antibodies, compared with the mice immunized with the antigen alone. In addition, IgM titers in the Muc1 + poly I:C group increased by 6.4 times, and IgG titers increased by 7.2 times, compared with the Muc1 group. Of note, ploy I:C altered the balance of IgG2a and IgG1, inducing more extensive production of IgG2a antibody, compared with the control group. It has been reported that IgG2a is directly associated with the T helper (Th)1 cell-mediated immune response, whereas IgG1 is associated with the Th2 immune response (30). This suggests that Muc1 + poly I:C may be involved in modulating cellular immunity, with positive effects in preventing tumor progression.

IFN-γ and IL-2 cytokines are increased in mice immunized with Muc1 + poly I:C or TLA + poly I:C

Cytokine patterns can reflect effector populations of immune cells to a certain degree. Th1 cytokines activate macrophages, natural killer cells and cellular immunity, whereas Th2 cytokines tend to promote humoral immune response (31). In the present study, at 10 days following the final immunization, splenocytes in the different treatment groups were stimulated with Muc1 at a concentration of 1 µg/ml, and the production of the IFN-γ and IL-2 Th1 cytokines were examined using an ELISA. As shown in Fig. 6A and B, the levels of IFN-γ and IL-2 in the splenocytes of the mice immunized with Muc1 + ploy I:C were significantly higher, as compared with those of the mice immunized with Muc1 alone (P<0.01). The present study also used TLAs from AtT-20 cells as antigens, and the results demonstrated that the levels of IFN-γ and IL-2 in the splenocytes of the mice immunized with TLA + poly I:C were significantly higher, as compared with those of the mice immunized with TLA alone (P<0.01).

Cytotoxic T cell (CTL) activity is increased following immunization with TLA + poly I:C, as compared with TLA alone

To demonstrate the cytotoxic activity of splenocytes from the immunized mice, the present study examined the release of cytosolic LDH into the culture medium by damaged AtT20 cells. At 10 days following the final immunization, damage to the membranes of the AtT20 cells was evaluated in a 24 h cytotoxicity assay by measuring LDH release. The LDH release assays were performed with splenocytes as effector cells and AtT20 cells as target cells. The E:T cell ratios were 5:1, 10:1 and 20:1. As shown in Fig. 7, the CTL response was significantly higher in the mice immunized with TLA + poly I:C, as compared with that in those the mice immunized with TLA alone (P<0.01) or with PBS (P<0.01), which demonstrated that TLA + poly I:C effectively induced cytotoxic activity to induce tumor cell death.

Discussion

Cancer can be treated using several techniques, including surgery, chemotherapy, radiation therapy, immunotherapy and mAb therapy. Cancer immunotherapy has emerged as one of the four most commonly used treatment modality, in addition to surgery, chemotherapy and radiotherapy (32). Vaccines have been investigated for a long time and have been used for the treatment of infectious disease or cancer (3335), however, progression has remained slower, compared with other forms of immunotherapy (3537). This is partly due to the weak immunogenicity of the tumor antigens. In the present study the, expression levels of Muc1 in pituitary tumors were examined, and the results demonstrated increased positive expression of Muc1 in invasive pituitary tumors. The aim of the present study was to induce potent antitumor T-cell responses to assist in the therapy of pituitary tumors. Thus, Muc1 was used as the tumor antigen and poly I:C, the ligand of TLR3, was selected as the adjuvant to prime the antitumor immune response (3740). The results of the present study demonstrated that Muc1 + poly I:C successfully induced effective antitumor immune responses.

DCs are considered to be important antigen presenting cells, and they are the most effective cells for antigen presentation and T cell stimulation (41). The maturation of DCs is important for the initiation of an adaptive immune response, thus, the present study detected the presence of certain major histocompatibility complex class II molecules and T-cell costimulatory molecules on the surface of PBMC-derived DCs. The results of the present study revealed that, Muc1 + poly I:C significantly enhanced the expression levels of CD40, CD80, CD83 and HLA-DR, in which CD83 is considered a relatively specific marker of mature DCs, and CD14 is a marker of monocytes. Treatment with Muc1 + poly I:C increased the expression of CD83 and decreased the expression of CD14, which demonstrated that Muc1 + poly I:C promoted the maturation of the DCs. Additionally, Muc1 + poly I:C stimulated the secretion of TNF-α, IL-6 and IL-1β cytokines, which may assist in attracting neutrophils, monocytes and DCs to inflammatory sites and induce the adaptive immune response (42,43).

It has been reported that the Th1-type immune response is important for protection against viral infections, tumor cells and several pathogens. Th1 cells can secret cytokines, including IFN-γ and IL-2, which activate macrophages and are involved in the generation of CTLs. In the present study, the mice injected with Muc1 + poly I:C showed higher levels of IFN-γ and IL-2 in the splenocytes when stimulated with Muc1. The CTL response following the final immunization was also examined in the different groups of mice. Supporting the idea that the induction of IFN-γ and IL-2 suggested polarization towards the Th1 response, higher specific CTL activity was detected when stimulated with Muc1 antigens in vitro.

In the mouse model, immunization with Muc1 + poly I:C induced more extensive production of anti-Muc1, compared with the antigen alone. Notable, activation of the Th1 immune response was marked, as was the switch in the balance of IgG1 and IgG2a, which may contribute to the shift between the Th2 and Th1 immune respons, and was positive for preventing the progression of pituitary tumors.

In conclusion, Muc1 + poly I:C stimulated the expression of costimulatory molecules and promoted the secretion of inflammatory cytokines, IL-6, TNF-α, IL-1β, by human PBMC-derived DCs. In the mouse model, immunization with Muc1 + poly I:C enhanced Muc1-specific antibody titers and induced the shift between the Th2 and Th1 immune response. Therefore, it may offer potential as a useful strategy for the treatment of malignant pituitary tumors.

Acknowledgments

This study was supported by the National Natural Science Foundation of China (grant no. 81171119).

References

1 

Loyo-Varela M, Herrada-Pineda T, Revilla-Pacheco F and Manrique-Guzman S: Pituitary tumor surgery: Review of 3004 cases. World Neurosurg. 79:331–336. 2013. View Article : Google Scholar

2 

Salehi F, Kovacs K, Scheithauer BW, Lloyd RV and Cusimano M: Pituitary tumor-transforming gene in endocrine and other neoplasms: A review and update. Endocr Relat Cancer. 15:721–743. 2008. View Article : Google Scholar : PubMed/NCBI

3 

Boutros C, Cheng-Robles D and Goldenkranz R: Intestinal neuroendocrine tumor in a patient with pituitary adenoma. A case report and review of the current screening recommendations. J Med Case Rep. 1:1402007. View Article : Google Scholar : PubMed/NCBI

4 

Sogani J, Yang W, Lavi E, Zimmerman RD and Gupta A: Sellar collision tumor involving metastatic lung cancer and pituitary adenoma: Radiologic-pathologic correlation and review of the literature. Clin Imaging. 38:318–321. 2014. View Article : Google Scholar : PubMed/NCBI

5 

Ironside JW: Best practice No 172: Pituitary gland pathology. J Clin Pathol. 56:561–568. 2003. View Article : Google Scholar : PubMed/NCBI

6 

Nistor RF: Parasellar classification of pituitary adenomas. Neurosurgery. 35:542–544. 1994.PubMed/NCBI

7 

Kovacs K, Horvath E and Vidal S: Classification of pituitary adenomas. J Neurooncol. 54:121–127. 2001. View Article : Google Scholar

8 

Sánchez-Tejada L, Sánchez-Ortiga R, Moreno-Pérez O, Montañana CF, Niveiro M, Tritos NA and Alfonso AM: Pituitary tumor transforming gene and insulin-like growth factor 1 receptor expression and immunohistochemical measurement of Ki-67 as potential prognostic markers of pituitary tumors aggressiveness. Endocrinol Nutr. 60:358–367. 2013. View Article : Google Scholar : PubMed/NCBI

9 

Matsuno A, Teramoto A, Takekoshi S, Sanno N, Osamura RY and Kirino T: Expression of plurihormonal mRNAs in somatotrophic adenomas detected using a nonisotopic in situ hybridization method: Comparison with lactotrophic adenomas. Hum Pathol. 26:272–279. 1995. View Article : Google Scholar : PubMed/NCBI

10 

Baskar R, Lee KA, Yeo R and Yeoh KW: Cancer and radiation therapy: Current advances and future directions. Int J Med Sci. 9:193–199. 2012. View Article : Google Scholar : PubMed/NCBI

11 

Bergmann-Leitner ES, Duncan EH and Leitner WW: Identification and targeting of tumor escape mechanisms: A new hope for cancer therapy? Curr Pharm Des. 9:2009–2023. 2003. View Article : Google Scholar : PubMed/NCBI

12 

Ahmed S, Pahwa P, Fields A, Chandra-Kanthan S, Iqbal N, Zaidi A, Reeder B, Plaza FA, Zhu T and Leis A: Predictive factors of the use of systemic therapy in stage iv colorectal cancer: Who gets chemotherapy? Oncology. 88:289–297. 2015. View Article : Google Scholar : PubMed/NCBI

13 

Voloshin T, Voest EE and Shaked Y: The host immunological response to cancer therapy: An emerging concept in tumor biology. Exp Cell Res. 319:1687–1695. 2013. View Article : Google Scholar : PubMed/NCBI

14 

Rini BI, Weinberg V, Fong L, Conry S, Hershberg RM and Small EJ: Combination immunotherapy with prostatic acid phosphatase pulsed antigen-presenting cells (provenge) plus bevacizumab in patients with serologic progression of prostate cancer after definitive local therapy. Cancer. 107:67–74. 2006. View Article : Google Scholar : PubMed/NCBI

15 

Thara E, Dorff TB, Pinski JK and Quinn DI: Vaccine therapy with sipuleucel-T (Provenge) for prostate cancer. Maturitas. 69:296–303. 2011. View Article : Google Scholar : PubMed/NCBI

16 

Mao Q, Li L, Zhang C, Sun Y, Liu S and Cui S: Clinical effects of immunotherapy of DC-CIK combined with chemotherapy in treating patients with metastatic breast cancer. Pak J Pharm Sci. 28(3 Suppl): 1055–1058. 2015.PubMed/NCBI

17 

Shi YJ, Ren HY, Cen XN, Dong YJ, Ma MX, Zhao YL, Zhu Y and Yu JR: Dendritic cells elicit cellular immune response by targeting to capture breast cancer cells. Zhonghua zhong liu za zhi. 30:107–111. 2008.In Chinese. PubMed/NCBI

18 

Chen W, Rains N, Young D and Stubbs RS: Dendritic cell-based cancer immunotherapy: Potential for treatment of colorectal cancer? J Gastroenterol Hepatol. 15:698–705. 2000. View Article : Google Scholar : PubMed/NCBI

19 

Carmi Y, Spitzer MH, Linde IL, Burt BM, Prestwood TR, Perlman N, Davidson MG, Kenkel JA, Segal E, Pusapati GV, et al: Allogeneic IgG combined with dendritic cell stimuli induce antitumour T-cell immunity. Nature. 521:99–104. 2015. View Article : Google Scholar : PubMed/NCBI

20 

Ahmed Ali HA, Di J, Mei W, Zhang YC, Li Y, Du ZW and Zhang GZ: Antitumor activity of lentivirus-mediated interleukin -12 gene modified dendritic cells in human lung cancer in vitro. Asian Pac J Cancer Prev. 15:611–616. 2014. View Article : Google Scholar : PubMed/NCBI

21 

Bilusic M, Heery C and Madan RA: Immunotherapy in prostate cancer: Emerging strategies against a formidable foe. Vaccine. 29:6485–6497. 2011. View Article : Google Scholar : PubMed/NCBI

22 

Pardoll D and Drake C: Immunotherapy earns its spot in the ranks of cancer therapy. J Exp Med. 209:201–0209. 2012. View Article : Google Scholar : PubMed/NCBI

23 

Corfield AP: Mucins: A biologically relevant glycan barrier in mucosal protection. Biochim Biophys Acta. 1850:236–252. 2015. View Article : Google Scholar

24 

Kao CJ, Wurz GT, Monjazeb AM, Vang DP, Cadman TB, Griffey SM, Wolf M and DeGregorio MW: Antitumor effects of cisplatin combined with tecemotide immunotherapy in a human MUC1 transgenic lung cancer mouse model. Cancer Immunol Res. 2:581–589. 2014. View Article : Google Scholar : PubMed/NCBI

25 

Pérez-Sánchez J, Estensoro I, Redondo MJ, Calduch-Giner JA, Kaushik S and Sitjà-Bobadilla A: Mucins as diagnostic and prognostic biomarkers in a fish-parasite model: Transcriptional and functional analysis. PLoS One. 8:e654572013. View Article : Google Scholar : PubMed/NCBI

26 

Asano R, Takemura S, Tsumoto K, Sakurai N, Teramae A, Ebara S, Katayose Y, Shinoda M, Suzuki M, Imai K, et al: Functional construction of the anti-mucin core protein (MUC1) antibody MUSE11 variable regions in a bacterial expression system. J Biochem. 127:673–679. 2000. View Article : Google Scholar : PubMed/NCBI

27 

Cao X, Zhang W, Wan T, He L, Chen T, Yuan Z, Ma S, Yu Y and Chen G: Molecular cloning and characterization of a novel CXC chemokine macrophage inflammatory protein-2 gamma chemoattractant for human neutrophils and dendritic cells. J Immunol. 165:2588–2595. 2000. View Article : Google Scholar : PubMed/NCBI

28 

Costa CI, Delgado IF, de Costa JA, de Carvalho RF, Mouta Sda S Jr, Vianna CO and de Moraes MT: Establishment and validation of an ELISA for the quantitation of HBsAg in recombinant hepatitis B vaccines. J Virol Methods. 172:32–037. 2011. View Article : Google Scholar

29 

Jones N, Agrawal D, Elrefaei M, Hanson A, Novitsky V, Wong JT and Cao H: Evaluation of antigen-specific responses using in vitro enriched T cells. J Immunol Methods. 274:139–147. 2003. View Article : Google Scholar : PubMed/NCBI

30 

Holdsworth SR, Kitching AR and Tipping PG: Th1 and Th2 T helper cell subsets affect patterns of injury and outcomes in glomerulonephritis. Kidney Int. 55:1198–1216. 1999. View Article : Google Scholar : PubMed/NCBI

31 

Muraille E and Leo O: Revisiting the Th1/Th2 paradigm. Scand J Immunol. 47:1–9. 1998. View Article : Google Scholar : PubMed/NCBI

32 

Mishra J, Drummond J, Quazi SH, Karanki SS, Shaw JJ, Chen B and Kumar N: Prospective of colon cancer treatments and scope for combinatorial approach to enhanced cancer cell apoptosis. Crit Rev Oncol Hematol. 86:232–250. 2013. View Article : Google Scholar :

33 

Kirkwood JM, Moschos S and Wang W: Strategies for the development of more effective adjuvant therapy of melanoma: Current and future explorations of antibodies, cytokines, vaccines, and combinations. Clin Cancer Res. 12(Suppl): S2331–S2336. 2006. View Article : Google Scholar

34 

Kirkwood JM, Strawderman MH, Ernstoff MS, Smith TJ, Borden EC and Blum RH: Interferon alfa-2b adjuvant therapy of high-risk resected cutaneous melanoma: The eastern cooperative oncology group trial EST 1684. J Clin Oncol. 14:7–17. 1996.PubMed/NCBI

35 

Terando A, Sabel MS and Sondak VK: Melanoma: Adjuvant therapy and other treatment options. Curr Treat Options Oncol. 4:187–199. 2003. View Article : Google Scholar : PubMed/NCBI

36 

Krishnan L, Deschatelets L, Stark FC, Gurnani K and Sprott GD: Archaeosome adjuvant overcomes tolerance to tumor-associated melanoma antigens inducing protective CD8 T cell responses. Clin Dev Immunol. 2010:5784322010. View Article : Google Scholar

37 

Mechl Z and Kopecny J: Current results with surgery and adjuvant chemotherapy in malignant melanoma. Arch Geschwulstforsch. 56:367–371. 1986.In German.

38 

Inao T, Harashima N, Monma H, Okano S, Itakura M, Tanaka T, Tajima Y and Harada M: Antitumor effects of cytoplasmic delivery of an innate adjuvant receptor ligand, poly (I:C), on human breast cancer. Breast Cancer Res Treat. 134:89–100. 2012. View Article : Google Scholar

39 

Huang YK, Zheng Z, Cheng CX, Wang LY, Li YR and Qiu F: The antitumor effect of the toll-like receptor 3 ligand polyinosinic-cytidylic acid as an adjuvant. Cancer Immunol Immunother. 62:237–244. 2013. View Article : Google Scholar

40 

Martinez-Gil L, Goff PH, Hai R, Garcia-Sastre A, Shaw ML and Palese P: A Sendai virus-derived RNA agonist of RIG-I as a virus vaccine adjuvant. J Virol. 87:1290–1300. 2013. View Article : Google Scholar :

41 

Ebner S, Ratzinger G, Krosbacher B, Schmuth M, Weiss A, Reider D, Kroczek RA, Herold M, Heufler C, Fritsch P and Romani N: Production of IL-12 by human monocyte-derived dendritic cells is optimal when the stimulus is given at the onset of maturation, and is further enhanced by IL-4. J Immunol. 166:633–641. 2001. View Article : Google Scholar

42 

Lindstedt M, Johansson-Lindbom B and Borrebaeck CA: Global reprogramming of dendritic cells in response to a concerted action of inflammatory mediators. Int Immunol. 14:1203–1213. 2002. View Article : Google Scholar : PubMed/NCBI

43 

Yang Y, Ochando J, Yopp A, Bromberg JS and Ding Y: IL-6 plays a unique role in initiating c-Maf expression during early stage of CD4 T cell activation. J Immunol. 174:2720–2729. 2005. View Article : Google Scholar : PubMed/NCBI

Related Articles

Journal Cover

April-2016
Volume 13 Issue 4

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

Sign up for eToc alerts

Recommend to Library

Copy and paste a formatted citation
x
Spandidos Publications style
Sui D, Ma L, Li M, Shao W, Du H, Li K, Li Z and Li Z: Mucin 1 and poly I:C activates dendritic cells and effectively eradicates pituitary tumors as a prophylactic vaccine. Mol Med Rep 13: 3675-3683, 2016
APA
Sui, D., Ma, L., Li, M., Shao, W., Du, H., Li, K. ... Li, Z. (2016). Mucin 1 and poly I:C activates dendritic cells and effectively eradicates pituitary tumors as a prophylactic vaccine. Molecular Medicine Reports, 13, 3675-3683. https://doi.org/10.3892/mmr.2016.4964
MLA
Sui, D., Ma, L., Li, M., Shao, W., Du, H., Li, K., Li, Z., Li, Z."Mucin 1 and poly I:C activates dendritic cells and effectively eradicates pituitary tumors as a prophylactic vaccine". Molecular Medicine Reports 13.4 (2016): 3675-3683.
Chicago
Sui, D., Ma, L., Li, M., Shao, W., Du, H., Li, K., Li, Z., Li, Z."Mucin 1 and poly I:C activates dendritic cells and effectively eradicates pituitary tumors as a prophylactic vaccine". Molecular Medicine Reports 13, no. 4 (2016): 3675-3683. https://doi.org/10.3892/mmr.2016.4964