Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 72
Filter
1.
Oncol Rep ; 42(5): 2057-2064, 2019 Nov.
Article in English | MEDLINE | ID: mdl-31545443

ABSTRACT

The interaction between tumor necrosis factor receptor superfamily, member 4 (OX40) on T cells and the OX40 ligand (OX40L) on antigen­presenting cells (APCs) is a pivotal step for T­cell activation and the promotion of antitumor immunity. However, it is hypothesized that soluble OX40 (sOX40) in blood suppresses T­cell activation by blocking the OX40/OX40L interaction. In the present study, the association between blood sOX40 levels and the clinical characteristics of advanced colorectal cancer (CRC) patients was investigated. Blood was collected from 22 patients with advanced CRC. Blood sOX40 levels were determined by enzyme­linked immunosorbent assay (ELISA). Messenger RNA (mRNA) expression encoding OX40 or cytokines was analyzed by quantitative RT­PCR. Blood sOX40 levels were positively correlated with the blood levels of carbohydrate antigen (CA) 19­9, carcinoembryonic antigen (CEA), C­reactive protein (CRP) and soluble programmed cell death ligand­1 (PD­L1) in patients but negatively correlated with the blood levels of albumin. Blood sOX40 levels were not correlated with the mRNA expression of interferon (IFN)­gamma, interleukin (IL)­6, IL­10 and IL­4 in the peripheral blood mononuclear cells (PBMCs) of the patients and were not correlated with the frequency of programmed cell death­1 (PD­1) expressing CD4+, CD8+ and CD56+ cells. Notably, according to both univariate and multivariate analyses, high blood sOX40 levels were significantly correlated with a reduced survival time in patients. Although activated Jurkat cells (a human T cell line) exhibited an upregulation of sOX40 production and OX40 mRNA expression, the OX40 mRNA expression of the PBMCs of patients was not correlated with blood sOX40 levels. High blood levels of sOX40 were correlated with a reduced survival time in patients with advanced CRC, possibly associated with the suppression of antitumor immunity by sOX40.


Subject(s)
Colorectal Neoplasms/mortality , Receptors, OX40/blood , Receptors, OX40/genetics , Up-Regulation , Adult , Aged , Aged, 80 and over , Antigens, Tumor-Associated, Carbohydrate/blood , C-Reactive Protein/metabolism , Carcinoembryonic Antigen/blood , Colorectal Neoplasms/genetics , Colorectal Neoplasms/immunology , Female , Gene Expression Regulation, Neoplastic , Humans , Jurkat Cells , Male , Middle Aged , Neoplasm Staging , Organothiophosphorus Compounds/blood , Serum Albumin, Human/metabolism , Survival Analysis
3.
Pancreatology ; 19(1): 88-96, 2019 Jan.
Article in English | MEDLINE | ID: mdl-30416041

ABSTRACT

BACKGROUND/OBJECTIVES: Pancreatic cancer consists of various subpopulations of cells, some of which have aggressive proliferative properties. The molecules responsible for the aggressive proliferation of pancreatic cancer may become molecular targets for the therapies against pancreatic cancer. METHODS: From a human pancreatic cancer cell line, MIA PaCa-2, MIA PaCa-2-A cells with an epithelial morphology and MIA PaCa-2-R cells with a non-epithelial morphology were clonogenically isolated by the limiting dilution method. Gene expression of these subpopulations was analyzed by DNA microarray. Gene knockdown was performed using siRNA. RESULTS: Although the MIA PaCa-2-A and MIA PaCa-2-R cells displayed the same DNA short tandem repeat (STR) pattern identical to that of the parental MIA PaCa-2 cells, the MIA PaCa-2-A cells were more proliferative than the MIA PaCa-2-R cells both in culture and in tumor xenografts generated in immunodeficient mice. Furthermore, the MIA PaCa-2-A cells were more resistant to gemcitabine than the MIA PaCa-2-R cells. DNA microarray analysis revealed a high expression of claudin (CLDN) 7 in the MIA PaCa-2-A cells, as opposed to a low expression in the MIA PaCa-2-R cells. The knockdown of CLDN7 in the MIA PaCa-2-A cells induced a marked inhibition of proliferation. The MIA PaCa-2-A cells in which CLDN7 was knocked down exhibited a decreased expression of phosphorylated extracellular signal-regulated kinase (p-Erk)1/2 and G1 cell cycle arrest. CONCLUSIONS: CLDN7 may be expressed in the rapidly proliferating and dominant cell population in human pancreatic cancer tissues and may be a novel molecular target for the treatment of pancreatic cancer.


Subject(s)
Claudins/metabolism , Pancreatic Neoplasms/drug therapy , Animals , Cell Line, Tumor , Cell Proliferation , Claudins/genetics , G1 Phase Cell Cycle Checkpoints , Gene Expression Regulation, Neoplastic , Gene Knockdown Techniques , Humans , Mice , Neoplasms, Experimental , Oligonucleotide Array Sequence Analysis , RNA, Small Interfering
4.
Cancer Immunol Immunother ; 67(9): 1477-1479, 2018 09.
Article in English | MEDLINE | ID: mdl-30084040

ABSTRACT

The graphs are incorrectly identified in Fig. 3i, s and should be replaced with the following.

5.
Clin Lung Cancer ; 19(5): 410-417.e1, 2018 09.
Article in English | MEDLINE | ID: mdl-29859759

ABSTRACT

BACKGROUND: Biomarkers for predicting the effect of anti-programmed cell death 1 (PD-1) monoclonal antibody against non-small-cell lung cancer (NSCLC) are urgently required. Although it is known that the blood levels of soluble programmed cell death ligand 1 (sPD-L1) are elevated in various malignancies, the nature of sPD-L1 has not been thoroughly elucidated. We investigated the significance of plasma sPD-L1 levels as a biomarker for anti-PD-1 monoclonal antibody, nivolumab therapy. PATIENTS AND METHODS: The present prospective study included 39 NSCLC patients. The patients were treated with nivolumab at the dose of 3 mg/kg every 2 weeks, and the effects of nivolumab on NSCLC were assessed according to the change in tumor size, time to treatment failure (TTF), and overall survival (OS). The baseline plasma sPD-L1 concentration was determined using an enzyme-linked immunosorbent assay. RESULTS: The area under the curve of the receiver operating characteristic curve was 0.761. The calculated optimal cutoff point for sPD-L1 in the plasma samples was 3.357 ng/mL. Of the 39 patients, 59% with low plasma sPD-L1 levels achieved a complete response or partial response and 25% of those with high plasma sPD-L1 levels did so. In addition, 22% of the patients with low plasma sPD-L1 levels developed progressive disease compared with 75% of those with high plasma sPD-L1 levels. The TTF and OS were significantly longer for those patients with low plasma sPD-L1 levels compared with the TTF and OS for those with high plasma sPD-L1 levels. CONCLUSION: The clinical benefit from nivolumab therapy was significantly associated with the baseline plasma sPD-L1 levels. Plasma sPD-L1 levels might represent a novel biomarker for the prediction of the efficacy of nivolumab therapy against NSCLC.


Subject(s)
B7-H1 Antigen/blood , Carcinoma, Non-Small-Cell Lung/blood , Lung Neoplasms/blood , Neoplasm Recurrence, Local/blood , Nivolumab/therapeutic use , Adenocarcinoma/blood , Adenocarcinoma/drug therapy , Adenocarcinoma/secondary , Aged , Aged, 80 and over , Antineoplastic Agents, Immunological/therapeutic use , Biomarkers, Tumor/blood , Carcinoma, Large Cell/blood , Carcinoma, Large Cell/drug therapy , Carcinoma, Large Cell/secondary , Carcinoma, Non-Small-Cell Lung/drug therapy , Carcinoma, Non-Small-Cell Lung/secondary , Carcinoma, Squamous Cell/blood , Carcinoma, Squamous Cell/drug therapy , Carcinoma, Squamous Cell/secondary , Female , Follow-Up Studies , Humans , Lung Neoplasms/drug therapy , Lung Neoplasms/pathology , Lymphatic Metastasis , Male , Middle Aged , Neoplasm Recurrence, Local/drug therapy , Neoplasm Recurrence, Local/pathology , Prognosis , Prospective Studies , Survival Rate
6.
J Cancer Res Clin Oncol ; 144(8): 1601-1611, 2018 Aug.
Article in English | MEDLINE | ID: mdl-29858682

ABSTRACT

PURPOSE: Metastasis of non-small cell lung cancer (NSCLC), indicating hematogenous dissemination, is more frequent in patients harboring epidermal growth factor receptor (EGFR) mutations, who respond dramatically to EGFR-tyrosine kinase inhibitors (TKIs). METHODS: Based on the proposed association of miliary pulmonary metastasis and EGFR mutations in the previous studies, we conducted a retrospective study to assess survival of NSCLC with miliary pulmonary metastases in 223 patients harboring EGFR mutations who were treated with single agent EGFR-TKIs. RESULTS: Progression-free survival (PFS) and overall survival (OS) with single agent EGFR-TKIs were 11.7 months [95% confidence interval (CI) 9.6-13.7] and 23.7 months (95% CI 20.3-26.9), respectively. Patients with and without miliary pulmonary metastases were matched using propensity scores (n = 29 per group) based on clinical characteristics. After matching, the PFS were 8.2 months (95% CI 5.2-15.0) and 14.3 months (95% CI 9.6-30.0) (p = 0.02) in patients with and without miliary pulmonary metastases, respectively. Conversely, the OS were 15.3 months (95% CI 10.6-19.4) and 27.9 months (95% CI 22.0-33.0) (p = 0.003) in patients with and without miliary pulmonary metastases, respectively. By multivariate analysis, miliary pulmonary metastasis was associated with poor prognosis (p = 0.0035). CONCLUSION: The prognosis of patients with advanced NSCLC harboring EGFR mutations with miliary pulmonary metastasis demonstrated significantly worse outcomes compared to those without miliary pulmonary metastasis.


Subject(s)
Carcinoma, Non-Small-Cell Lung/genetics , Carcinoma, Non-Small-Cell Lung/mortality , ErbB Receptors/genetics , Lung Neoplasms/genetics , Lung Neoplasms/mortality , Mutation , Adult , Aged , Aged, 80 and over , Carcinoma, Non-Small-Cell Lung/drug therapy , Carcinoma, Non-Small-Cell Lung/enzymology , Disease-Free Survival , Female , Humans , Lung Neoplasms/drug therapy , Lung Neoplasms/enzymology , Male , Middle Aged , Protein Kinase Inhibitors/therapeutic use , Retrospective Studies
7.
Cancer Immunol Res ; 6(3): 320-331, 2018 03.
Article in English | MEDLINE | ID: mdl-29358173

ABSTRACT

We investigated the efficacy of a Wilms' tumor gene 1 (WT1) vaccine combined with gemcitabine (GEMWT1) and compared it with gemcitabine (GEM) monotherapy for advanced pancreatic ductal adenocarcinoma (PDAC) in a randomized phase II study. We randomly assigned HLA-A*02:01- or HLA-A*24:02-positive patients with advanced PDAC to receive GEMWT1 or GEM. We assessed WT1-specific immune responses via delayed-type hypersensitivity (DTH) to the WT1 peptide and a tetramer assay to detect WT1-specific cytotoxic T lymphocytes (WT1-CTL). Of 91 patients enrolled, 85 were evaluable (GEMWT1: n = 42; GEM: n = 43). GEMWT1 prolonged progression-free survival [PFS; hazard ratio (HR), 0.66; P = 0.084] and improved overall survival rate at 1 year (1-year OS%; GEMWT1: 35.7%; GEM: 20.9%). However, the difference in OS was not significant (HR: 0.82; P = 0.363). These effects were particularly evident in metastatic PDAC (PFS: HR 0.51, P = 0.0017; 1-year OS%: GEMWT1 27.3%; GEM 11.8%). The combination was well tolerated, with no unexpected serious adverse events. In patients with metastatic PDAC, PFS in the DTH-positive GEMWT1 group was significantly prolonged, with a better HR of 0.27 compared with the GEM group, whereas PFS in the DTH-negative GEMWT1 group was similar to that in the GEM group (HR 0.86; P = 0.001). DTH positivity was associated with an increase in WT1-CTLs induced by the WT1 vaccine. GEM plus the WT1 vaccine prolonged PFS and may improve 1-year OS% in advanced PDAC. These clinical effects were associated with the induction of WT1-specific immune responses. Cancer Immunol Res; 6(3); 320-31. ©2018 AACR.


Subject(s)
Adenocarcinoma/therapy , Antimetabolites, Antineoplastic/therapeutic use , Cancer Vaccines/therapeutic use , Carcinoma, Pancreatic Ductal/therapy , Deoxycytidine/analogs & derivatives , Vaccines, Subunit/therapeutic use , WT1 Proteins , Adult , Aged , Combined Modality Therapy , Deoxycytidine/therapeutic use , Female , Humans , Male , Middle Aged , Progression-Free Survival , Gemcitabine
8.
Int Immunopharmacol ; 54: 39-45, 2018 Jan.
Article in English | MEDLINE | ID: mdl-29100036

ABSTRACT

Programmed cell death ligand-1 (PD-L1) plays a pivotal role in the suppression of antitumour immunity by binding to programmed cell death-1 (PD-1) on tumouricidal cytotoxic T lymphocytes (CTLs), rendering them inactive. As blockade of PD-1/PD-L1 interaction by the monoclonal antibodies induced effective T cell-mediated antitumour response, suppression of PD-L1 expression in tumour cells by the chemical agent might contribute to treatment against malignant tumours. Nafamostat mesilate (NM), a serine protease inhibitor that is frequently used in the clinic, potently suppressed interferon-gamma (IFN-gamma)-induced up-regulation of PD-L1 in cultured human lung cancer cells (HLC-1) at both the messenger RNA (mRNA) and protein levels. Interestingly, suppression of IFN-gamma-induced up-regulation of human leukocyte antigen (HLA)-ABC by NM was limited, suggesting that NM did not block CTL responses to tumour cells. NM treatment did not affect the activation status of signal transducer and activator of transcription (STAT) 1 or the induction of interferon regulatory factor (IRF)-1 expression in IFN-gamma-treated HLC-1 cells. Although NM treatment promoted the phosphorylation of extracellular signal-regulated kinases (Erk) 1/2, an Erk inhibitor, U0126, could not reverse the suppression of PD-L1 up-regulation by IFN-gamma. Suppression of IFN-gamma-induced up-regulation of PD-L1 by NM was not associated with the inhibition of nuclear factor kappa B (NF-kB) or protease-activated receptor (PAR)-1 pathway. Besides HLC-1 cells, NM suppressed IFN-gamma-induced PD-L1 up-regulation in three human pancreatic cancer cell lines. NM could potentiate the antitumour effect of cancer vaccines or immune checkpoint inhibitors by preventing IFN-gamma-induced PD-L1 up-regulation and blocking immune checkpoint suppression.


Subject(s)
Adenocarcinoma/drug therapy , B7-H1 Antigen/metabolism , CD8-Positive T-Lymphocytes/immunology , Guanidines/pharmacology , Lung Neoplasms/drug therapy , Pancreatic Neoplasms/drug therapy , Serine Proteinase Inhibitors/pharmacology , B7-H1 Antigen/genetics , Benzamidines , Cancer Vaccines , Cell Line, Tumor , HLA Antigens/metabolism , Humans , Immunosuppression Therapy , Interferon-gamma/metabolism , Up-Regulation
9.
Cancer Immunol Immunother ; 67(3): 495-505, 2018 Mar.
Article in English | MEDLINE | ID: mdl-29243049

ABSTRACT

BACKGROUND: The status of antitumor immunity represented by the expression of programmed cell death-1 (PD-1)/programmed cell death-ligand 1 (PD-L1) and immune cell (IC) infiltration is unknown in HIV-infected patients with non-small cell lung cancer (NSCLC). METHODS: Fifteen HIV-infected patients with NSCLC were compared with 29 non-HIV-infected patients with NSCLC. Analysis of 13 propensity-score-matched patients in the two groups was also compared. The expression of PD-1/PD-L1 and tumor infiltration by CD4+, CD8+, and CD56+ immune cells were examined by immunohistochemistry; score of ≥ 2 was defined as positive. RESULTS: Although high PD-L1 expression in tumor cells was observed in HIV and non-HIV cohorts, the association of PD-1/PD-L1 was significant only in the HIV cohort. In overall as well as the propensity-matched analyses, HIV-infected patients with high PD-L1 expression showed shorter survival than HIV-infected patients with low PD-L1 expression; no significant difference was observed in this respect in the non-HIV cohort. CONCLUSION: High PD-L1 expression in tumor tissue was associated with poor prognosis in HIV-infected NSCLC patients but not in non-HIV-infected NSCLC patients. These results suggest that antitumor immunity by PD-1/PD-L1 axis might be suppressed more in HIV-infected NSCLC patients as compared to their non-HIV-infected counterparts.


Subject(s)
Adenocarcinoma/pathology , B7-H1 Antigen/metabolism , Carcinoma, Non-Small-Cell Lung/pathology , Carcinoma, Squamous Cell/pathology , HIV Infections/immunology , Lung Neoplasms/pathology , Lymphocytes, Tumor-Infiltrating/immunology , Adenocarcinoma/immunology , Adenocarcinoma/metabolism , Adult , Aged , Carcinoma, Non-Small-Cell Lung/immunology , Carcinoma, Non-Small-Cell Lung/metabolism , Carcinoma, Squamous Cell/immunology , Carcinoma, Squamous Cell/metabolism , Case-Control Studies , Female , Follow-Up Studies , HIV/immunology , HIV Infections/complications , Humans , Lung Neoplasms/immunology , Lung Neoplasms/metabolism , Male , Middle Aged , Prognosis , Survival Rate
10.
PLoS One ; 12(11): e0188934, 2017.
Article in English | MEDLINE | ID: mdl-29190754

ABSTRACT

Adjuvants are indispensable for achieving a sufficient immune response from vaccinations. From a functional viewpoint, adjuvants are classified into two categories: "physical adjuvants" increase the efficacy of antigen presentation by antigen-presenting cells (APC) and "signal adjuvants" induce the maturation of APC. Our previous study has demonstrated that a physical adjuvant can be encrypted into proteinous antigens by creating artificial proteins from combinatorial assemblages of epitope peptides and those peptide sequences having propensities to form certain protein structures (motif programming). However, the artificial antigens still require a signal adjuvant to maturate the APC; for example, co-administration of the Toll-like receptor 4 (TLR4) agonist monophosphoryl lipid A (MPLA) was required to induce an in vivo immunoreaction. In this study, we further modified the previous artificial antigens by appending the peptide motifs, which have been reported to have agonistic activity for TLR4, to create "adjuvant-free" antigens. The created antigens with triple TLR4 agonistic motifs in their C-terminus have activated NF-κB signaling pathways through TLR4. These proteins also induced the production of the inflammatory cytokine TNF-α, and the expression of the co-stimulatory molecule CD40 in APC, supporting the maturation of APC in vitro. Unexpectedly, these signal adjuvant-encrypted proteins have lost their ability to be physical adjuvants because they did not induce cytotoxic T lymphocytes (CTL) in vivo, while the parental proteins induced CTL. These results confirmed that the manifestation of a motif's function is context-dependent and simple addition does not always work for motif-programing. Further optimization of the molecular context of the TLR4 agonistic motifs in antigens should be required to create adjuvant-free antigens.


Subject(s)
Antigen-Presenting Cells/cytology , Toll-Like Receptor 4/agonists , Animals , Antigen-Presenting Cells/immunology , Cell Line , Female , Humans , Immunity, Cellular , Mice , Mice, Inbred C57BL
11.
Oncol Rep ; 37(4): 1943-1952, 2017 Apr.
Article in English | MEDLINE | ID: mdl-28260012

ABSTRACT

Specimens obtained with endoscopic ultrasound-guided fine needle aspiration (EUS-FNA) are often tiny and fragmented leading to an inconclusive and doubtful diagnosis. To overcome the limitations of EUS-FNA in the cytological diagnosis of pancreatic adenocarcinoma (PCA), we evaluated whether quantification of the S100P protein combined with EUS-FNA reliably discriminated between PCA and benign pancreatic lesions (BPL). A high sensitivity sandwich ELISA for S100P protein was developed to aid in the detection of PCA in small samples obtained using EUS-FNA. After experimental verification of the sandwich ELISA with cell lines and mouse xenograft tumors, 27 consecutive patients with suspicious PCA who underwent EUS-FNA were enrolled in the present study examining the combination of S100P protein assessment and EUS-FNA cytology. The concentration of the S100P protein in EUS-FNA samples from the PCA group was significantly higher than that in the BPL group (P=0.04). Using receiver operating characteristic curve analysis, we determined the S100P protein cut-off value for PCA diagnosis to be 99.8 ng/ml. The S100P protein levels combined with EUS-FNA cytology to detect PCA showed the following diagnostic values: sensitivity, 94.4% [95% confidence interval (CI), 75.7-99.1%]; specificity, 88.9% (95% CI, 51.8-99.7%); positive predictive value, 94.4% (95% CI, 72.7-99.9%); negative predictive value, 88.9% (95% CI, 51.8-99.7%); accuracy, 92.6% (95% CI, 75.7­99.1%); and area under the curve, 0.92 (95% CI, 0.79-1.00). We established a novel quantitative analysis for the S100P protein in EUS-FNA samples which, when combined with EUS-FNA cytology, could provide promising results for the reliable diagnosis of PCA.


Subject(s)
Adenoma/diagnosis , Calcium-Binding Proteins/biosynthesis , Cytodiagnosis , Neoplasm Proteins/biosynthesis , Neoplasms/diagnosis , Pancreatic Neoplasms/diagnosis , Adenoma/genetics , Adenoma/pathology , Adult , Aged , Aged, 80 and over , Animals , Calcium-Binding Proteins/genetics , Cell Line, Tumor , Endoscopic Ultrasound-Guided Fine Needle Aspiration , Female , Gene Expression Regulation, Neoplastic , Humans , Male , Mice , Middle Aged , Neoplasm Proteins/genetics , Neoplasms/genetics , Neoplasms/pathology , Pancreatic Neoplasms/genetics , Pancreatic Neoplasms/pathology , Pilot Projects , Xenograft Model Antitumor Assays
12.
Lung Cancer ; 104: 1-6, 2017 02.
Article in English | MEDLINE | ID: mdl-28212990

ABSTRACT

OBJECTIVES: Programmed cell death-ligand 1 (PD-L1) expressed in tumor tissues is a key molecule for immune suppression, given its role in immune checkpoints. The significance and implication of soluble PD-L1 (sPD-L1) in the blood of lung cancer patients remain unknown. PATIENTS AND METHODS: Blood samples were prospectively collected from patients with advanced lung cancer, and the plasma sPD-L1 concentrations were measured by enzyme-linked immunosorbent assay. The correlations of the plasma sPD-L1 levels with clinico-pathological status, laboratory data, and survival of the patients were analyzed. RESULTS: Ninety-six patients with advanced lung cancer were analyzed, including 73 with adenocarcinoma, 12 with squamous cell carcinoma, and seven with small-cell lung cancer. Sixty-five were naïve to chemotherapy, and 20 had received two or more lines of chemotherapy. The mean plasma sPD-L1 concentration of all the patients was 6.95±2.90ng/ml (range 2.30-20.0ng/ml), and this value is significantly increased compared with that previously reported for normal subjects. No correlation of the plasma sPD-L1 level with histological subtypes, adenocarcinoma genetic status, smoking history, clinical stage or laboratory data was found. However, overall survival was significantly reduced in patients with high (≥7.32ng/ml) compared with low (<7.32ng/ml) plasma sPD-L1 levels (13.0 vs. 20.4 months, p=0.037). Multivariate analysis revealed that high sPD-L1 levels were significantly related to poor prognosis (hazard ratio 1.99, p=0.041). CONCLUSION: High plasma sPD-L1 levels were associated with poor prognosis in patients with advanced lung cancer, possibly associated with suppression of anti-tumor immunity. Clinical trial register and their clinical registration number: UMIN%000014760.


Subject(s)
B7-H1 Antigen/blood , Immune Tolerance/immunology , Lung Neoplasms/metabolism , Prognosis , Survival , Tumor Escape/immunology , Adenocarcinoma/genetics , Adenocarcinoma/pathology , Adult , Aged , Aged, 80 and over , B7-H1 Antigen/metabolism , Biomarkers, Tumor/blood , Female , Humans , Lung Neoplasms/drug therapy , Lung Neoplasms/mortality , Lung Neoplasms/pathology , Male , Middle Aged , Prospective Studies
13.
Cancer Immunol Immunother ; 65(12): 1499-1509, 2016 12.
Article in English | MEDLINE | ID: mdl-27688162

ABSTRACT

BACKGROUND: This trial was designed to evaluate the safety and clinical responses to a combination of temozolomide (TMZ) chemotherapy and immunotherapy with fusions of DCs and glioma cells in patients with glioblastoma (GBM). METHOD: GBM patients were assigned to two groups: a group of recurrent GBMs after failing TMZ-chemotherapy against the initially diagnosed glioma (Group-R) or a group of newly diagnosed GBMs (Group-N). Autologous cultured glioma cells obtained from surgical specimens were fused with autologous DCs using polyethylene glycol. The fusion cells (FC) were inoculated intradermally in the cervical region. Toxicity, progression-free survival (PFS), and overall survival (OS) of this trial were evaluated. Expressions of WT-1, gp-100, and MAGE-A3, recognized as chemoresistance-associated peptides (CAP), were confirmed by immunohistochemistry of paraffin-embedded tumor samples. Patient's PBMCs of pre- and post-vaccination were evaluated by tetramer and ELISPOT assays. RESULTS: FC-immunotherapy was well tolerated in all patients. Medians of PFS and OS of Group-R (n = 10) were 10.3 and 18.0 months, and those of Group-N (n = 22) were 18.3 and 30.5 months, respectively. Up-regulation and/or cytoplasmic accumulation of CAPs was observed in the recurrent tumors of Group-R patients compared with their initially excised tumors. Specific immune responses against CAPs were observed in the tetramer and ELISPOT assays. CONCLUSIONS: The combination of TMZ-treatment leading to up-regulation and/or cytoplasmic accumulation of CAPs, with FC-immunotherapy as a means of producing specific immunity against CAPs, may safely induce anti-tumor effects in patients with GBM.


Subject(s)
Antineoplastic Agents, Alkylating/therapeutic use , Dacarbazine/analogs & derivatives , Dendritic Cells/immunology , Glioblastoma/drug therapy , Glioblastoma/immunology , Glioma/immunology , Immunotherapy/methods , Adult , Aged , Antineoplastic Agents, Alkylating/administration & dosage , Dacarbazine/administration & dosage , Dacarbazine/therapeutic use , Female , Humans , Male , Middle Aged , Temozolomide , Up-Regulation
14.
Cancer Sci ; 107(5): 682-9, 2016 May.
Article in English | MEDLINE | ID: mdl-26920337

ABSTRACT

Although rituximab, a chimeric monoclonal antibody that specifically binds to CD20, has significantly improved the prognosis for diffuse large B cell lymphoma (DLBCL), one-third of DLBCL patients demonstrate resistance to rituximab or relapse after rituximab treatment. Thus, a novel approach to rituximab-based treatment is likely to be required to improve the efficacy of DLBCL treatment. As complement dependent cytotoxicity (CDC) is a key mechanism mediating rituximab's tumoricidal activity, rituximab binding to CD20 on tumor cells is a critical factor for effective rituximab-based treatments against DLBCL. We found that gemcitabine (GEM), but not lenalidomide (LEN) or azacitidine (AZA), can upregulate CD20 expression in TK and KML-1 cells, two human DLBCL cell lines. Treatment of TK and KML-1 cells with GEM enhanced CD20 expression at both the mRNA and protein levels. CD20 upregulation by GEM treatment was accompanied by increased rituximab binding to CD20. In TK cells, GEM treatment synergistically increased rituximab-mediated CDC activity in a dose-dependent manner. In KML cells, GEM treatment also induced upregulation of complement regulatory proteins, possibly leading to resistance to CDC. Treatment with LEN, a drug that did not upregulate CD20, did not enhance rituximab-mediated CDC activity. GEM treatment activated nuclear factor-kappa B (NF-kB) signaling in these cells. Furthermore, a specific inhibitor to NF-kB suppressed GEM-induced CD20 upregulation, indicating that GEM-induced NF-kB activation is closely associated with CD20 upregulation. These results suggest that when used in combination, GEM might enhance the antitumor efficacy of rituximab against DLBCL due to its unique ability to upregulate CD20.


Subject(s)
Antigens, CD20/metabolism , Complement System Proteins/drug effects , Cytotoxicity, Immunologic/drug effects , Deoxycytidine/analogs & derivatives , Lymphoma, B-Cell/immunology , Lymphoma, B-Cell/metabolism , Rituximab/pharmacology , Up-Regulation/drug effects , Azacitidine/pharmacology , Cell Line, Tumor , Complement System Proteins/immunology , Deoxycytidine/pharmacology , Humans , Lenalidomide , Lymphoma, B-Cell/drug therapy , Lymphoma, Large B-Cell, Diffuse/drug therapy , Lymphoma, Large B-Cell, Diffuse/metabolism , NF-kappa B/metabolism , Thalidomide/analogs & derivatives , Thalidomide/pharmacology , Gemcitabine
15.
World J Gastrointest Pharmacol Ther ; 7(1): 133-8, 2016 Feb 06.
Article in English | MEDLINE | ID: mdl-26855819

ABSTRACT

"Vaccell" is a dendritic cell (DC)-based cancer vaccine which has been established in Japan. The DCs play central roles in deciding the direction of host immune reactions as well as antigen presentation. We have demonstrated that DCs treated with a streptococcal immune adjuvant OK-432, produce interleukin-12, induce Th1-dominant state, and elicit anti-tumor effects, more powerful than those treated with the known DC-maturating factors. We therefore decided to mature DCs by the OK-432 for making an effective DC vaccine, Vaccell. The 255 patients with inoperable pancreatic cancer who received standard chemotherapy combined with DC vaccines, were analyzed retrospectively. Survival time of the patients with positive delayed type hypersensitivity (DTH) skin reaction was significantly prolonged as compared with that of the patients with negative DTH. The findings strongly suggest that there may be "Responders" for the DC vaccine in advanced pancreatic cancer patients. We next conducted a small-scale prospective clinical study. In this trial, we pulsed HLA class II-restricted WT1 peptide (WT1-II) in addition to HLA class I-restricted peptide (WT1-I) into the DCs. Survival of the patients received WT1-I and -II pulsed DC vaccine was significantly extended as compared to that of the patients received DCs pulsed with WT1-I or WT1-II alone. Furthermore, WT1-specific DTH positive patients showed significantly improved the overall survival as well as progression-free survival as compared to the DTH negative patients. The activation of antigen-specific immune responses by DC vaccine in combination with standard chemotherapy may be associated with a good clinical outcome in advanced pancreatic cancer. We are now planning a pivotal study of the Vaccell in appropriate protocols in Japan.

16.
PLoS One ; 10(12): e0142981, 2015.
Article in English | MEDLINE | ID: mdl-26642349

ABSTRACT

Chondroitin sulfate E (CS-E), a highly sulfated glycosaminoglycan, is known to promote tumor invasion and metastasis. Because the presence of CS-E is detected in both tumor and stromal cells in pancreatic ductal adenocarcinoma (PDAC), multistage involvement of CS-E in the development of PDAC has been considered. However, its involvement in the early stage of PDAC progression is still not fully understood. In this study, to clarify the direct role of CS-E in tumor, but not stromal, cells of PDAC, we focused on carbohydrate sulfotransferase 15 (CHST15), a specific enzyme that biosynthesizes CS-E, and investigated the effects of the CHST15 siRNA on tumor cell proliferation in vitro and growth in vivo. CHST15 mRNA is highly expressed in the human pancreatic cancer cell lines PANC-1, MIA PaCa-2, Capan-1 and Capan-2. CHST15 siRNA significantly inhibited the expression of CHST15 mRNA in these four cells in vitro. Silencing of the CHST15 gene in the cells was associated with significant reduction of proliferation and up-regulation of the cell cycle inhibitor-related gene p21CIP1/WAF1. In a subcutaneous xenograft tumor model of PANC-1 in nude mice, a single intratumoral injection of CHST15 siRNA almost completely suppressed tumor growth. Reduced CHST15 protein signals associated with tumor necrosis were observed with the treatment with CHST15 siRNA. These results provide evidence of the direct action of CHST15 on the proliferation of pancreatic tumor cells partly through the p21CIP1/WAF1 pathway. Thus, CHST15-CS-E axis-mediated tumor cell proliferation could be a novel therapeutic target in the early stage of PDAC progression.


Subject(s)
Cell Proliferation/genetics , Gene Silencing/physiology , Membrane Glycoproteins/genetics , Pancreatic Neoplasms/genetics , Sulfotransferases/genetics , Adenocarcinoma/genetics , Animals , Carcinoma, Pancreatic Ductal/genetics , Cell Cycle/genetics , Cell Line, Tumor , Disease Progression , Gene Expression Regulation, Neoplastic/genetics , Humans , Mice , Mice, Inbred BALB C , Mice, Nude , RNA, Messenger/genetics , RNA, Small Interfering/genetics , Up-Regulation/genetics , Xenograft Model Antitumor Assays/methods
17.
BMC Cancer ; 15: 726, 2015 Oct 16.
Article in English | MEDLINE | ID: mdl-26475267

ABSTRACT

BACKGROUND: Although pancreatic ductal adenocarcinomas (PDAs) widely express HER2, the expression level is generally low. If HER2 expression in PDA cells could be enhanced by treatment with a given agent, then combination therapy with that agent and trastuzumab emtansine (T-DM1), a chemotherapeutic agent that is a conjugate of trastuzumab, might lead to significant antitumor effects against PDA. METHODS: Cell proliferation was examined by spectrophotometry. HER2 expression was examined by flow cytometry, immunoblot and quantitative reverse transcription polymerase chain reaction. T-DM1 binding to cells was examined by flow cytometry and enzyme-linked immunosorbent assay. RESULTS: Out of 5 tested human PDA cell lines, including MIA PaCa-2, three showed increases in HER2 expression after gemcitabine (GEM) treatment. The binding of T-DM1 to GEM-treated MIA PaCa-2 cells was higher than to untreated MIA PaCa-2 cells. Treatment with GEM and T-DM1 showed synergic cytotoxic effects on MIA PaCa-2 cells in vitro. Cells in the G2M phase of the cell cycle were retained after GEM treatment and showed higher levels of HER2 expression, possibly contributing to the synergic effect of GEM and T-DM1. CONCLUSIONS: Combined treatment with GEM and T-DM1 might confer a potent therapeutic modality against PDA as a result of GEM-mediated HER2 up-regulation.


Subject(s)
Adenocarcinoma/drug therapy , Cell Proliferation/drug effects , Pancreatic Neoplasms/drug therapy , Receptor, ErbB-2/biosynthesis , Adenocarcinoma/genetics , Adenocarcinoma/pathology , Ado-Trastuzumab Emtansine , Animals , Antibodies, Monoclonal, Humanized/administration & dosage , Antineoplastic Combined Chemotherapy Protocols/administration & dosage , Cell Line, Tumor , Deoxycytidine/administration & dosage , Deoxycytidine/analogs & derivatives , Gene Expression Regulation, Neoplastic/drug effects , Humans , Maytansine/administration & dosage , Maytansine/analogs & derivatives , Mice , Pancreatic Neoplasms/genetics , Pancreatic Neoplasms/pathology , Receptor, ErbB-2/genetics , Trastuzumab , Xenograft Model Antitumor Assays , Gemcitabine
18.
Oncol Rep ; 34(4): 2099-105, 2015 Oct.
Article in English | MEDLINE | ID: mdl-26239720

ABSTRACT

Monoclonal antibody therapy for immune checkpoint blockade has achieved promising results for several types of malignant tumors. For the future treatment of gastrointestinal stromal tumors (GISTs) by immune checkpoint blockade, expression of immune checkpoint-related molecules that suppress antitumor immunity in GISTs was examined. Infiltration of immune cell types into 19 GIST tissues was analyzed by immunohistochemistry, and expression of T cell immunoglobulin and mucin protein 3 (Tim-3) and programmed cell death-1 (PD-1) in the infiltrated immune cells was examined by immunofluorescence microscopy. The expression status of galectin-9 in the GIST tumor cells was also determined by immunohistochemistry. All the GIST tissues showed CD8+ T cell infiltration and 8 showed CD56+ natural killer (NK) cell infiltration, and the numbers of infiltrated CD8+ T and NK cells were strongly correlated. However, these CD8+ T and NK cells were CD69-negative inactivated cells. Tim-3 was expressed in the infiltrated NK cells in 6/8 (75%) of the GIST tissues. Expression of galectin-9, a ligand of Tim-3, was observed in 13/19 (68.4%) GIST tissues and all of the GIST tissues with Tim-3+ NK cell infiltration showed positive galectin-9 expression. No PD-1 expression in the infiltrated NK cells and neither Tim-3 nor PD-1 expression was observed in the infiltrated CD8+ T cells. Interaction between Tim-3 in infiltrated NK cells and galectin-9 in tumor cells may be involved in an immune checkpoint mechanism for suppression of antitumor immunity in GISTs. Blockade of the Tim-3/galectin-9 pathway may become a new strategy for GIST treatment.


Subject(s)
Galectins/metabolism , Gastrointestinal Stromal Tumors/immunology , Killer Cells, Natural/immunology , Membrane Proteins/metabolism , Adult , Aged , Aged, 80 and over , Antigens, CD/metabolism , Antigens, Differentiation, T-Lymphocyte/metabolism , CD56 Antigen/metabolism , CD8-Positive T-Lymphocytes/immunology , Female , Gastrointestinal Stromal Tumors/metabolism , Hepatitis A Virus Cellular Receptor 2 , Humans , Lectins, C-Type/metabolism , Male , Middle Aged
19.
Oncol Rep ; 34(1): 504-10, 2015 Jul.
Article in English | MEDLINE | ID: mdl-25976081

ABSTRACT

Trastuzumab emtansine (T-DM1), trastuzumab-conjugated with a cytotoxic agent, has shown promising antitumor effects in breast cancer. Since a good therapeutic response using T-DM1 treatment requires high human epidermal growth factor receptor 2 (HER2) expression, breast cancers with low or no HER2 expression have not been used for T-DM1 treatment. The aim of the present study was to show that treatment of low HER2-expressing breast cancer cells with gemcitabine (GEM) enhanced HER2 expression using RT-qPCR, immunoblot and flow cytometric analysis. The results showed that GEM treatment significantly enhanced HER2 expression in MDA-MB-231, MCF7 and BT-20 breast cancer cells, while paclitaxel (PTX) treatment induced lower or no enhancement in HER2 expression. The expression of HER2 mRNA was also enhanced in GEM-treated MCF7 cells. Treatment with an inhibitor for nuclear factor-(NF)-κB suppressed GEM-induced HER2 upregulation, indicating that NF-κB activation by GEM may be associated with HER2 upregulation. T-DM1 binding to HER2 on MCF-7 cells was enhanced by GEM pretreatment and the combined treatment of GEM and T-DM1 synergistically inhibited the proliferation of MCF7 cells. Thus, the combined treatment with GEM and T-DM1 may be a promising therapeutic modality for low HER2-expressing breast cancers, which was facilitated by the unique HER2-upregulating effect of GEM.


Subject(s)
Antibodies, Monoclonal, Humanized/pharmacology , Antineoplastic Agents/pharmacology , Breast Neoplasms/drug therapy , Deoxycytidine/analogs & derivatives , Maytansine/analogs & derivatives , Receptor, ErbB-2/genetics , Up-Regulation , Ado-Trastuzumab Emtansine , Breast Neoplasms/genetics , Breast Neoplasms/metabolism , Cell Line, Tumor , Cell Proliferation/drug effects , Deoxycytidine/pharmacology , Drug Synergism , Female , Humans , MCF-7 Cells , Maytansine/pharmacology , Paclitaxel/pharmacology , Receptor, ErbB-2/metabolism , Trastuzumab , Gemcitabine
20.
Int J Oncol ; 46(6): 2679, 2015 06.
Article in English | MEDLINE | ID: mdl-25846893

ABSTRACT

Some errors in Fig. 3B and Fig. 6C have been identified. The errors do not change the conclusion of the paper. The conclusion is supported by other figures in the paper, as well as results described in the text. The corrected Fig. 3B and Fig. 6C are shown below. [the original article was published in the International Journal of Oncology 45: 470-478, 2014 DOI: 10.3892/ijo.2014.2433]

SELECTION OF CITATIONS
SEARCH DETAIL
...