Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 27
Filter
1.
J Formos Med Assoc ; 2024 Mar 16.
Article in English | MEDLINE | ID: mdl-38494360

ABSTRACT

BACKGROUND: Perioperative immunosuppressants, such as surgical stress and opioid use may downregulate anti-cancer immunocytes for patients undergoing pancreatectomy. Thoracic epidural analgesia (TEA) may attenuate these negative effects and provide better anti-cancer immunocyte profile change than intravenous analgesia using opioid. METHODS: We randomly assigned 108 adult patients undergoing pancreatectomy to receive one of two 72-h postoperative analgesia protocols: one was TEA, and the other was intravenous patient-controlled analgesia (IV-PCA). The perioperative proportional changes of immunocytes relevant to anticancer immunity-namely natural killer (NK) cells, cytotoxic T cells, helper T cells, mature dendritic cells, and regulatory T (Treg) cells were determined at 1 day before surgery, at the end of surgery and on postoperative day 1,4 and 7 using flow cytometry. In addition, the progression-free survival and overall survival between the two groups were compared. RESULTS: After surgery, the proportions of NK cells and cytotoxic T cells were significantly decreased; the proportion of B cells and mature dendritic cells and Treg cells were significantly increased. However, the proportions of helper T cells exhibited no significant change. These results were comparable between the two groups. Furthermore, there were no significant differences in progression-free survival (52.75 [39.96] and 57.48 [43.66] months for patients in the TEA and IV-PCA groups, respectively; p = 0.5600) and overall survival (62.71 [35.48] and 75.11 [33.10] months for patients in the TEA and IV-PCA groups, respectively; p = 0.0644). CONCLUSIONS: TEA was neither associated with favorable anticancer immunity nor favorable oncological outcomes for patients undergoing pancreatectomy.

2.
J Formos Med Assoc ; 123(2): 228-237, 2024 Feb.
Article in English | MEDLINE | ID: mdl-37596109

ABSTRACT

BACKGROUND/PURPOSE: Efforts were made to explore the influence of diagnostic timing for cancer-associated thromboembolic events on survival of ovarian cancer patients. METHODS: We reviewed the medical records of 75 ovarian cancer patients with thromboembolism and evaluated the prognostic factors affecting disease-free survival and overall survival. RESULTS: These 75 patients were classified into two categories by the diagnostic timing of the thromboembolism, during (33 cases) and after (42 cases) initial diagnosis of ovarian cancer groups. The diagnostic timing of thromboembolism was not related to disease-free survival or overall survival of the studied population. Advanced disease stage, clear cell histology, interval debulking surgery, no recurrence/persistence of ovarian cancer, and patients treated with anticoagulant(s) treatment >3 months were associated with the disease-free survival. Advanced disease stage, clear cell histology, body mass index (BMI) ≥24 kg/m2 at the diagnosis of ovarian cancer, and no recurrence/persistence of ovarian cancer influenced the overall survival. In the subgroup analysis, compared to the after initial ovarian cancer diagnosis group, patients with stage I/II disease, BMI <24 kg/m2 at the diagnosis of ovarian cancer, or primary debulking surgery in the during cancer diagnosis group had longer disease-free survival, and overall survival benefit was observed in cases with stage I/II disease, or primary debulking surgery. CONCLUSION: The diagnostic timing of thromboembolism was not related to disease-free or overall survival of ovarian cancer patients, but associated with that of specific patient subgroups.


Subject(s)
Ovarian Neoplasms , Thromboembolism , Humans , Female , Ovarian Neoplasms/complications , Ovarian Neoplasms/diagnosis , Disease-Free Survival , Progression-Free Survival , Anticoagulants/therapeutic use , Thromboembolism/etiology
3.
J Immunother Cancer ; 11(7)2023 07.
Article in English | MEDLINE | ID: mdl-37463789

ABSTRACT

BACKGROUND: Expression of immune checkpoints in the tumor microenvironment is one mechanism underlying paclitaxel (PTX) chemoresistance. This study aimed to investigate whether the addition of checkpoint blockade to PTX can improve the therapeutic efficacy against apparently disseminated intraperitoneal tumors. METHODS: We analyzed the in vivo expression of various immune checkpoints in CD3+CD8+ cytotoxic T cells from tumor-bearing mice treated with or without PTX and validated the tumor-killing activities of selected checkpoint-expressing T-cell subpopulations ex vivo. The regulation of selected checkpoints was investigated in vitro. The therapeutic effects of inhibition of a targeted checkpoint pathway with antibodies added to PTX therapy were examined. RESULTS: CD3+CD8+ T cells expressed with herpes virus entry mediator (HVEM), programmed cell death 1 (PD-1), and T-cell immunoglobulin domain and mucin domain 3 (TIM-3) in tumor-bearing hosts treated with PTX had effective tumoricidal activities. In addition to PTX and cytokines, B and T lymphocyte attenuator (BTLA) or homologous to lymphotoxin, exhibits inducible expression and competes with herpes simplex virus (HSV) glycoprotein D for binding to HVEM, a receptor expressed on T lymphocytes (LIGHT) interacting with HVEM can regulate the expression of PD-1 on CD3+CD8+ T cells. Interleukin (IL)-15 increased the percentage of HVEMhighgranzyme B (GZMB)+ cells among CD3+CD8+ T cells, which was suppressed by the BTLA/HVEM signal. LIGHT induced the percentage of HVEM+GZMB+ cells but not HVEMhighGZMB+ cells among CD3+CD8+ T cells. Expression of IL-15, BTLA, or LIGHT was detected in CD19+ B cells and regulated by damage-associated molecular patterns/Toll-like receptor interactions. In the tumor-bearing hosts treated with PTX, certain proportions of BTLA+ B or PD-1+ T lymphocytes were still noted. When dual inhibition of BTLA and PD-1 was added to PTX, the antitumor effects on intraperitoneally disseminated tumors can be significantly improved. CONCLUSIONS: Dual blockade of BTLA on B cells and PD-1 on cytotoxic T cells may have clinical potential for enhancing the efficacy of PTX in the treatment of tumors with intraperitoneal spread, including epithelial ovarian carcinomas.


Subject(s)
CD8-Positive T-Lymphocytes , Neoplasms , Mice , Animals , CD8-Positive T-Lymphocytes/metabolism , Programmed Cell Death 1 Receptor/metabolism , Receptors, Immunologic/metabolism , Cytokines/metabolism , Neoplasms/metabolism , Tumor Microenvironment
4.
Cancer Lett ; 523: 149-161, 2021 12 28.
Article in English | MEDLINE | ID: mdl-34606928

ABSTRACT

Radiation is an integral part of cancer therapy. With the emergence of oncolytic vaccinia virus immunotherapy, it is important to study the combination of radiation and vaccinia virus in cancer therapy. In this study, we investigated the anti-tumor effect of and immune mechanisms underlying the combination of high-dose hypofractionated stereotactic body radiotherapy (SBRT) and oncolytic vaccinia virus in preclinical murine models. The combination enhanced the in vivo anti-tumor effect and increased the numbers of splenic CD4+Ki-67+ helper T lymphocytes and CD8+Ki-67+ cytotoxic T lymphocytes. Combinational therapy also increased tumor-infiltrating CD3+CD4+ helper T lymphocytes and CD3+CD8+ cytotoxic T lymphocytes, but decreased tumor-infiltrating regulatory T cells. In addition, SBRT combined with oncolytic vaccinia virus enhanced in vitro cell death, partly through necroptosis, and subsequent release of damage-associated molecular patterns (DAMPs), and shifted the macrophage M1/M2 ratio. We concluded that SBRT combined with oncolytic vaccinia virus can trigger tumor cell necroptosis and modify macrophages through the release of DAMPs, and then generate potent anti-tumor immunity and effects. Thus, combined therapy is potentially an important strategy for clinical cancer therapy.


Subject(s)
Alarmins/physiology , Necroptosis/physiology , Neoplasms, Experimental/therapy , Oncolytic Virotherapy , Radiosurgery , Animals , CD8-Positive T-Lymphocytes/immunology , Cell Line, Tumor , Combined Modality Therapy , Female , Macrophages/physiology , Mice , Mice, Inbred C57BL , Neoplasms, Experimental/immunology , Neoplasms, Experimental/pathology , Tumor Microenvironment , Vaccinia virus
5.
J Immunother Cancer ; 7(1): 313, 2019 11 21.
Article in English | MEDLINE | ID: mdl-31753019

ABSTRACT

BACKGROUND: The standard treatment for epithelial ovarian carcinoma (EOC) is surgery followed by platinum/paclitaxel-based chemotherapy, but the overall survival rate is poor. The purpose of this study was to investigate the therapeutic potential of chemotherapy combined with inhibition of B and T lymphocyte attenuator (BTLA) for clinical use to treat EOC. METHODS: Initially, we evaluated the potential application of chemotherapy combined with anti-BTLA antibody in an animal model. We then analyzed the distribution and regulation of BTLA expression on immunocytes in vitro. Finally, we examined the correlation between BTLA expression levels in cancerous tissues and prognosis in 254 EOC cases. RESULTS: The combination of chemotherapy and anti-BTLA antibody for inhibiting BTLA significantly reduced peritoneal tumor volume and extended survival in tumor-bearing mice. In addition, BTLA could be identified mostly on B lymphocytes, especially on CD19hi B cells, rather than on T lymphocytes and natural killer cells. Under regulation of interleukins 6 and 10, more BTLA+CD19hi B lymphocytes could be induced through AKT and STAT3 signaling pathways. Detectable BTLA expression in ovarian cancerous tissues was associated with worse disease-free and overall survivals of EOC patients. CONCLUSIONS: BTLA detected in cancerous tissues can predict poor outcome of EOC patients. Inhibition of BTLA combined with chemotherapy can elevate immune activation and generate potent anti-tumor effects. Thus, the combination of chemotherapy and anti-BTLA antibody may hold potential clinical application for the treatment of EOC patients. TRIAL REGISTRATION: The Trial Registration Number was NCT00854399.


Subject(s)
Antibodies, Monoclonal/therapeutic use , Carcinoma, Ovarian Epithelial/drug therapy , Ovarian Neoplasms/drug therapy , Receptors, Immunologic/antagonists & inhibitors , Adult , Animals , Antibodies, Monoclonal/pharmacology , Antigens, CD19/immunology , B-Lymphocytes/drug effects , B-Lymphocytes/immunology , Carcinoma, Ovarian Epithelial/immunology , Carcinoma, Ovarian Epithelial/mortality , Cytokines/immunology , Female , Humans , Mice, Inbred C57BL , Middle Aged , Ovarian Neoplasms/immunology , Ovarian Neoplasms/mortality , Prognosis , Proportional Hazards Models , Receptors, Immunologic/immunology
6.
Cancers (Basel) ; 11(9)2019 Sep 19.
Article in English | MEDLINE | ID: mdl-31546897

ABSTRACT

The immuno-inhibitory checkpoint PD-L1, regulated by tumor cells and antigen-presenting cells (APCs), dampened the activation of T cells from the PD-1/PD-L1 axis. PD-L1-expressing APCs rather than tumor cells demonstrated the essential anti-tumor effects of anti-PD-L1 monotherapy in preclinical tumor models. Using the murine tumor model, we investigated whether anti-PD-L1 antibody increased the antigen-specific immune response and anti-tumor effects induced by the antigen-specific protein vaccine, as well as the possible mechanisms regarding activation of APCs. Anti-PD-L1 antibody combined with the PEK protein vaccine generated more potent E7-specific immunity (including the number and cytotoxic activity of E7-specific cytotoxic CD8+ T lymphocytes) and anti-tumor effects than protein vaccine alone. Anti-PD-L1 antibody enhanced the maturation of dendritic cells and the proportion of M1-like macrophages in tumor-draining lymph nodes and tumors in tumor-bearing mice treated with combinatorial therapy. PD-L1 blockade overturned the immunosuppressive status of the tumor microenvironment and then enhanced the E7 tumor-specific antigen-specific immunity and anti-tumor effects generated by an E7-specific protein vaccine through modulation of APCs in an E7-expressing small tumor model. Tumor-specific antigen (like HPV E7 antigen)-specific immunotherapy combined with APC-targeting modality by PD-L1 blockade has a high translational potential in E7-specific cancer therapy.

7.
J Formos Med Assoc ; 118(12): 1616-1622, 2019 Dec.
Article in English | MEDLINE | ID: mdl-31377113

ABSTRACT

BACKGROUND/PURPOSE: Oral cancer patients who survive for more than 5 years are supposed to have a reduced local cancer recurrence rate and survive longer. This study evaluated whether oral cancer patients who survived for more than 5 years might have reduced rates of local cancer recurrence, development of the second or third primary oral cancer, or the late regional cervical lymph node metastasis. METHODS: This study analyzed the clinical outcomes of 127 oral cancer patients (101 men and 26 women; mean age, 50.8 ± 12.1 years) who survived for more than 5 years after proper treatments of the initial primary oral cancers. RESULTS: The 127 primary oral cancers included 117 squamous cell carcinomas (SCCs), 7 mucoepidermoid carcinomas, and 3 others. Of the 127 oral cancer patients who survived for more than 5 years, 47 survived for 5-9 years, 45 for 10-14 years, 22 for 15-19 years, 10 for 20-24 years, two for 25-29 years, and one for 30 years. Ten patients had local cancer recurrence 5.4 years-13.7 years, 12 patients had a second or a third primary oral cancer 3.6 years-17.2 years, and one mucoepidermoid carcinoma patient had a late regional cervical lymph node metastasis 11.9 years after total excision of the initial primary oral cancers. CONCLUSION: Oral cancer patients who survive for more than 5 years may still have local cancer recurrence, the second or third primary oral cancer, or the late regional cervical lymph node metastasis but with a reduced rate.


Subject(s)
Carcinoma, Mucoepidermoid/mortality , Carcinoma, Squamous Cell/mortality , Mouth Neoplasms/mortality , Neoplasm Recurrence, Local/mortality , Neoplasms, Second Primary/mortality , Adolescent , Adult , Aged , Aged, 80 and over , Child , Female , Humans , Lymphatic Metastasis , Male , Middle Aged , Mouth Neoplasms/surgery , Neoplasm Staging , Survival Rate , Taiwan , Time Factors , Young Adult
8.
Cancers (Basel) ; 11(5)2019 May 02.
Article in English | MEDLINE | ID: mdl-31052575

ABSTRACT

The life span of dendritic cells (DCs) can become short following induced activation, which is associated with metabolic transition due to the regulation of mechanistic target of rapamycin (mTOR). The purpose of this study was to investigate the potential of inhibiting mTOR to modulate DC functions for elevating the anti-tumor effects of DNA vaccines. Therefore, the influences of various inhibitors of mTOR (mTORi) on the expressions of DC maturation markers, the abilities of antigen presenting and processing of BMM-derived DCs and the tumor killing effects of E7-specific CD8+ T lymphocytes activated by BMM-derived DCs were in vitro examined. The anti-tumor effects of connective tissue growth factor (CTGF)/E7 DNA vaccine and/or mTORi were also in vivo analyzed. In our study, suppressive effects of mTORi on the DC maturation markers expressed on BMMCs could be reversed. The mTORi-treated mature BMM-derived DCs tended to be non-apoptotic. These mTORi-treated BMM-derived DCs could have better antigen presenting and processing abilities. The E7-specific cytotoxic CD8+ T lymphocytes could have more potent tumoricidal activity following activation of mTORi-treated BMM-derived DCs. For tumor-bearing mice, those treated with CTGF/E7 DNA vaccine and mTORi indeed can have higher percentages of mature DCs in the TME, better disease control and longer survivals. Consequently, application of mTORi can be a pharmacological approach for temporally increasing life span, antigen presenting and antigen processing of DCs to strengthen the therapeutic outcome of cancer immunotherapy.

9.
Endocr Relat Cancer ; 26(1): 73-88, 2019 01 01.
Article in English | MEDLINE | ID: mdl-30121622

ABSTRACT

The role of chitinase-3-like protein 1 (CHI3L1) in ovarian cancer and the possible mechanisms were elucidated. CHI3L1 is a secreted glycoprotein and associated with inflammation, fibrosis, asthma, extracellular tissue remodeling and solid tumors. Our previous study showed CHI3L1 could be a potential prognostic biomarker for epithelial ovarian cancer and could protect cancer cells from apoptosis. Therefore, clinical data and quantitation of CHI3L1 of ovarian cancer patients, tumor spheroid formation, side-population assays, Aldefluor and apoptotic assays, ELISA, RT-PCR, immunoblotting and animal experiments were performed in two ovarian cancer cells lines, OVCAR3 and CA5171, and their CHI3L1-overexpressing and -knockdown transfectants. High expression of CHI3L1 was associated with poor outcome and chemoresistance in ovarian cancer patients. The mRNA expression of CHI3L1 in CA5171 ovarian cancer stem-like cells was 3-fold higher than in CA5171 parental cells. CHI3L1 promoted the properties of ovarian cancer stem-like cells including generating more and larger tumor spheroids and a higher percentage of ALDH+ in tumor cells and promoting resistance to cytotoxic drug-induced apoptosis. CHI3L1 could induce both the Akt (essential) and Erk signaling pathways, and then enhance expression of ß-catenin followed by SOX2, and finally promote tumor spheroid formation and other properties of ovarian cancer stem-like cells. OVCAR3 CHI3L1-overexpressing transfectants were more tumorigenic in vivo, whereas CA5171 CHI3L1-knockdown transfectants were not tumorigenic in vivo. CHI3L1 critically enhances the properties of ovarian cancer stem-like cells. CHI3L1 or CHI3L1-regulated signaling pathways and molecules could be potential therapeutic targets in ovarian cancer.


Subject(s)
Chitinase-3-Like Protein 1 , Ovarian Neoplasms/pathology , Animals , Cell Line, Tumor , Chitinase-3-Like Protein 1/genetics , Chitinase-3-Like Protein 1/metabolism , Female , Humans , MAP Kinase Signaling System , Mice, SCID , Middle Aged , Neoplastic Stem Cells , Ovarian Neoplasms/genetics , Ovarian Neoplasms/metabolism , Prognosis , Proto-Oncogene Proteins c-akt/metabolism , SOXB1 Transcription Factors/metabolism , Spheroids, Cellular , beta Catenin/metabolism
10.
Cancer Lett ; 444: 20-34, 2019 03 01.
Article in English | MEDLINE | ID: mdl-30543813

ABSTRACT

We determined the anti-tumor effects and possible mechanisms of an antigen-specific DNA vaccine combined with PD-1 or CTLA-4 blockade. Using the HPV16 E6/E7+ syngeneic mouse tumor model, we investigated whether anti-CTLA-4 antibody (Ab) or anti-PD-1 Ab increases the antigen-specific anti-tumor effects and immune response induced by CTGF/E7 chimeric DNA vaccine and the possible mechanisms. Anti-PD-1 Ab or anti-CTLA-4 Ab combined with E7-specific DNA vaccine generated more potent antigen-specific immunity, including anti-E7 Abs and the number and cytotoxic activity of E7-specific cytotoxic CD8+ T lymphocytes, and anti-tumor effects than E7-specific DNA vaccine alone. In addition, the number of systemic and intratumoral Tregs was lower with the anti-PD-1 or anti-CTLA-4 Ab and E7-specific DNA vaccine. Furthermore, anti-PD-1 and anti-CTLA-4 Abs could enhance the maturation and abilities of intratumoral DCs to activate E7-specific cytotoxic CD8+ T cells. Immune checkpoint blockade overcomes the immunosuppressive status of the tumor-microenvironment to enhance the antigen-specific immunity and anti-tumor effects generated by an antigen-specific DNA vaccine. Antigen-specific immunotherapy combined with immune checkpoint blockade can be a novel strategy in clinical cancer therapy.


Subject(s)
Antibodies, Monoclonal/pharmacology , CTLA-4 Antigen/immunology , Dendritic Cells/immunology , Neoplasms/immunology , Programmed Cell Death 1 Receptor/immunology , T-Lymphocytes, Cytotoxic/immunology , T-Lymphocytes, Regulatory/immunology , Animals , Antigens, Neoplasm/immunology , Apoptosis , CD8-Positive T-Lymphocytes/immunology , CTLA-4 Antigen/antagonists & inhibitors , Cell Proliferation , Female , Humans , Immunotherapy , Mice , Mice, Inbred C57BL , Neoplasms/pathology , Neoplasms/therapy , Programmed Cell Death 1 Receptor/antagonists & inhibitors , Tumor Cells, Cultured , Tumor Microenvironment , Vaccines, DNA/administration & dosage , Vaccines, DNA/immunology , Xenograft Model Antitumor Assays
11.
Medicine (Baltimore) ; 97(39): e12644, 2018 Sep.
Article in English | MEDLINE | ID: mdl-30278591

ABSTRACT

The ankle-brachial index (ABI) is a noninvasive method for screening for peripheral artery disease (PAD). However, false-negative findings of the ABI may limit its clinical use. The percentage of mean arterial pressure (%MAP) calculated from pulse volume recording has been reported to predict all-cause mortality. We hypothesized that the %MAP would be helpful to screen for PAD in subjects with a normal ABI. We examined whether using a combination of the ABI and %MAP would provide greater diagnostic sensitivity for PAD than using the ABI alone.In this cross-sectional study, we retrospectively reviewed the medical records of patients who had undergone multiple detector computed tomography (MDCT) angiography of the lower extremities following measurement of the ABI with pulse volume recording. PAD was diagnosed based on MDCT angiography.A total of 215 lower extremities of 114 patients were included in our analyses. An optimal cut-off %MAP value of 42.5% was used to diagnose PAD based on MDCT in patients with an ABI > 0.90. Using a combination of an ABI < 0.90 and a %MAP ≥ 42.5% as diagnostic criteria for PAD resulted in better sensitivity (76.9%) than using the ABI alone (56.5% for an ABI < 0.90 and 63.4% for an ABI < 1.00). Using logistic regression analysis, we found that patients having both an ABI < 0.90 and an ABI > 0.90 with a %MAP ≥ 42.5% had a significantly higher risk of PAD than those having an ABI > 0.90 with a %MAP < 42.5% (odds ratio = 7.165, P = .006; odds ratio = 12.544, P < .001; respectively).Both the sensitivity and specificity were better when using a combination of an ABI ≤ 0.90 and a %MAP ≥ 42.5% than when using a low or borderline ABI. The %MAP is helpful for PAD screening in subjects with an ABI > 0.90.


Subject(s)
Ankle Brachial Index , Arterial Pressure , Blood Pressure Determination/methods , Peripheral Arterial Disease/diagnosis , Aged , Aged, 80 and over , Case-Control Studies , Cross-Sectional Studies , Female , Humans , Male , Middle Aged , Multidetector Computed Tomography , Peripheral Arterial Disease/diagnostic imaging , Peripheral Arterial Disease/physiopathology , Retrospective Studies , Sensitivity and Specificity
12.
Epigenomics ; 10(11): 1397-1413, 2018 11.
Article in English | MEDLINE | ID: mdl-30324802

ABSTRACT

AIM: To investigate the CDH1, DLEC1 and SFRP5 gene methylation panel for advanced epithelial ovarian carcinoma (EOC). MATERIALS & METHODS: One hundred and seventy-seven advanced EOC specimens were evaluated by methylation-specific PCR. We also used The Cancer Genome Atlas dataset to evaluate the panel. RESULTS: The presence of two or more methylated genes was significant in recurrence (hazard ratio [HR]: 1.91 [1.33-2.76]; p = 0.002) and death (HR: 1.96 [1.26-3.06]; p = 0.006) in our cohort. In The Cancer Genome Atlas dataset, the presence of two or three methylated genes was significant in death (HR: 1.59 [1.15-2.18]; p = 0.0047) and close to the significance level in recurrence (HR: 1.37 [0.99-1.88]; p = 0.058). CONCLUSION: The CDH1, DLEC1 and SFRP5 methylation panel is a potential prognostic biomarker for advanced EOC.


Subject(s)
Antigens, CD/genetics , Biomarkers, Tumor/genetics , Cadherins/genetics , Carcinoma, Ovarian Epithelial/genetics , DNA Methylation , Eye Proteins/genetics , Membrane Proteins/genetics , Ovarian Neoplasms/genetics , Tumor Suppressor Proteins/genetics , Adaptor Proteins, Signal Transducing , Adult , Aged , Carcinoma, Ovarian Epithelial/pathology , Female , Humans , Middle Aged , Ovarian Neoplasms/pathology , Survival Analysis
13.
Cancer Lett ; 425: 152-163, 2018 07 01.
Article in English | MEDLINE | ID: mdl-29596890

ABSTRACT

As a tumor antigen, mesothelin (MSLN) can be identified in various malignancies. MSLN is potential for antigen-specific cancer vaccines. We generated a novel chimeric DNA vaccine using antigen-specific connective tissue growth factor lined with MSLN (CTGF/MSLN). The anti-tumor effects of the CTGF/MSLN DNA vaccine combined with anti-CD40 Ab and toll-like receptor 3 ligand-poly(I:C) were validated in an MSLN-expressing model. CTGF/MSLN DNA with anti-CD40Ab and poly(I:C) vaccinated mice demonstrated potent anti-tumor effects with longer survival and less tumor volumes. An increase in MSLN-specific CD8+ T cells and anti-MSLN Ab titers was also noted in CTGF/MSLN DNA with anti-CD40Ab and poly(I:C) vaccinated mice. The CTGF/MSLN DNA vaccine combined with immuno-modulator EGCG also generated potent anti-tumor effects. Immuno-modulators could enhance the antigen-specific anti-tumor effects of CTGF/MSLN DNA vaccine through promoting the DC maturation. In addition, MSLN-specific cell-based vaccine with AAV-IL-12 and the CTGF/MSLN DNA vaccine with anti-CD40Ab/polyp(I:C) generated more potent anti-tumor effects than the other combinational regimens. The results indicate that an MSLN-specific DNA vaccine combined with immuno-modulators may be an effective immunotherapeutic strategy to control MSLN-expressing tumors including ovarian and pancreastic cancers, and malignant mesothelioma.


Subject(s)
Antibodies, Monoclonal/administration & dosage , Catechin/analogs & derivatives , Connective Tissue Growth Factor/genetics , GPI-Linked Proteins/genetics , Lung Neoplasms/drug therapy , Mesothelioma/drug therapy , Poly I-C/administration & dosage , Vaccines, DNA/administration & dosage , Animals , Antibodies, Monoclonal/immunology , Antineoplastic Combined Chemotherapy Protocols/immunology , Cancer Vaccines/administration & dosage , Cancer Vaccines/immunology , Catechin/administration & dosage , Catechin/immunology , Cell Line, Tumor , Dendritic Cells/immunology , Drug Synergism , Female , Lung Neoplasms/immunology , Mesothelin , Mesothelioma/immunology , Mesothelioma, Malignant , Mice , Poly I-C/immunology , Treatment Outcome , Vaccines, DNA/immunology , Xenograft Model Antitumor Assays
14.
Mol Ther ; 26(2): 404-419, 2018 02 07.
Article in English | MEDLINE | ID: mdl-29248428

ABSTRACT

Ionizing radiation therapy is a well-established method of eradicating locally advanced tumors. Here, we examined whether local RT enhanced the potency of an antigen-specific DNA vaccine, and we investigated the possible underlying mechanism. Using the HPV16 E6/E7+ syngeneic TC-1 tumor, we evaluated the combination of CTGF/E7 vaccination with local irradiation with regard to synergistic antigen-specific immunity and anti-tumor effects. Tumor-bearing mice treated with local RT (6 Gy twice weekly) and CTGF/E7 DNA vaccination exhibited dramatically increased numbers of E7-specific CD8+ cytotoxic T cell precursors, higher titers of anti-E7 Abs, and significantly reduced tumor size. The combination of local RT and CTGF/E7 vaccination also elicited abscopal effects on non-irradiated local subcutaneous and distant pulmonary metastatic tumors. Local irradiation induced the expression of high-mobility group box 1 protein (HMGB-1) in apoptotic tumor cells and stimulated dendritic cell (DC) maturation, consequently inducing antigen-specific immune responses. Additionally, local irradiation eventually increased the effector-to-suppressor cell ratio in the tumor microenvironment. Overall, local irradiation enhanced the antigen-specific immunity and anti-tumor effects on local and distant metastatic tumors generated by an antigen-specific DNA vaccine. These findings suggest that the combination of irradiation with antigen-specific immunotherapy is a promising new clinical strategy for cancer therapy.


Subject(s)
Antigens, Neoplasm/immunology , Immunotherapy , Neoplasms/immunology , Neoplasms/pathology , Tumor Microenvironment/immunology , Animals , CD8-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/metabolism , Cancer Vaccines/immunology , Combined Modality Therapy , Cytotoxicity, Immunologic , Dendritic Cells/immunology , Dendritic Cells/metabolism , Disease Models, Animal , HMGB1 Protein/genetics , HMGB1 Protein/metabolism , Humans , Immunotherapy/methods , Mice , Neoplasms/genetics , Neoplasms/therapy , T-Lymphocytes, Regulatory/immunology , T-Lymphocytes, Regulatory/metabolism , Tumor Microenvironment/genetics , Whole-Body Irradiation , Xenograft Model Antitumor Assays
15.
Oncotarget ; 6(37): 39740-55, 2015 Nov 24.
Article in English | MEDLINE | ID: mdl-26452028

ABSTRACT

We propose CHI3L1 as a prognostic biomarker for patients with epithelial ovarian carcinoma (EOC) and also suggest possible biological functions of CHI3L1. We measured CHI3L1 expression with quantitative real time-polymerase chain reaction (qRT-PCR) in 180 women with EOC and evaluated correlations between CHI3L1 expression, clinicopathological characteristics, and the outcomes of the patients. The expression of CHI3L1 was higher in cancerous tissues than in normal tissues. The expression of CHI3L1 was also higher in patients with a serous histological type, advanced stage, and chemoresistance. Patients with high CHI3L1 expression had a shorter progression-free survival (p < 0.001)and overall survival (p < 0.001). Patients with high CHI3L1 expression also had a high risk of recurrence (p < 0.001)and death (p < 0.001). In vitro studies showed that CHI3L1 up-regulated the expression of anti-apoptotic Mcl-1 protein and hampered paclitaxel-induced apoptosis of ovarian cancer cells. These results suggest that CHI3L1 shows potential as a prognostic biomarker for EOC. CHI3L1 may promote chemoresistance via inhibition of drug-induced apoptosis by up-regulating Mcl-1.


Subject(s)
Adipokines/genetics , Drug Resistance, Neoplasm/genetics , Gene Expression Regulation, Neoplastic , Lectins/genetics , Neoplasms, Glandular and Epithelial/genetics , Ovarian Neoplasms/genetics , Adipokines/metabolism , Adult , Aged , Analysis of Variance , Apoptosis/drug effects , Apoptosis/genetics , Biomarkers, Tumor/genetics , Biomarkers, Tumor/metabolism , Blotting, Western , Carcinoma, Ovarian Epithelial , Cell Line, Tumor , Chitinase-3-Like Protein 1 , Disease-Free Survival , Female , Humans , Lectins/metabolism , Middle Aged , Myeloid Cell Leukemia Sequence 1 Protein/genetics , Myeloid Cell Leukemia Sequence 1 Protein/metabolism , Neoplasms, Glandular and Epithelial/metabolism , Neoplasms, Glandular and Epithelial/therapy , Outcome Assessment, Health Care/methods , Outcome Assessment, Health Care/statistics & numerical data , Ovarian Neoplasms/metabolism , Ovarian Neoplasms/therapy , Paclitaxel/pharmacology , Prognosis , Proportional Hazards Models , RNA Interference , Reverse Transcriptase Polymerase Chain Reaction
16.
Endocr Relat Cancer ; 22(5): 703-11, 2015 Oct.
Article in English | MEDLINE | ID: mdl-26150382

ABSTRACT

Aside from tumor cells, ovarian cancer-related ascites contains the immune components. The aim of this study was to evaluate whether a combination of clinical and immunological parameters can predict survival in patients with ovarian cancer. Ascites specimens and medical records from 144 ovarian cancer patients at our hospital were used as the derivation group to select target clinical and immunological factors to generate a risk-scoring system to predict patient survival. Eighty-two cases from another hospital were used as the validation group to evaluate this system. The surgical status and expression levels of interleukin 17a (IL17a) and IL21 in ascites were selected for the risk-scoring system in the derivation group. The areas under the receiver operating characteristic (AUROC) curves of the overall score for disease-free survival (DFS) of the ovarian cancer patients were 0.84 in the derivation group, 0.85 in the validation group, and 0.84 for all the patients. The AUROC curves of the overall score for overall survival (OS) of cases were 0.78 in the derivation group, 0.76 in the validation group, and 0.76 for all the studied patients. Good correlations between overall risk score and survival of the ovarian cancer patients were demonstrated by sub-grouping all participants into four groups (P for trend <0.001 for DFS and OS). Therefore, acombination of clinical and immunological parameters can provide a practical scoring system to predict the survival of patients with ovarian carcinoma. IL17a and IL21 can potentially be used as prognostic and therapeutic biomarkers.


Subject(s)
Adenocarcinoma, Clear Cell/pathology , Adenocarcinoma, Mucinous/pathology , Cystadenocarcinoma, Serous/pathology , Endometrial Neoplasms/pathology , Interleukin-17/metabolism , Interleukins/metabolism , Ovarian Neoplasms/pathology , Adenocarcinoma, Clear Cell/metabolism , Adenocarcinoma, Clear Cell/mortality , Adenocarcinoma, Clear Cell/surgery , Adenocarcinoma, Mucinous/metabolism , Adenocarcinoma, Mucinous/mortality , Adenocarcinoma, Mucinous/surgery , Biomarkers, Tumor/metabolism , Cystadenocarcinoma, Serous/metabolism , Cystadenocarcinoma, Serous/mortality , Cystadenocarcinoma, Serous/surgery , Endometrial Neoplasms/metabolism , Endometrial Neoplasms/mortality , Endometrial Neoplasms/surgery , Enzyme-Linked Immunosorbent Assay , Female , Follow-Up Studies , Humans , Middle Aged , Neoplasm Grading , Neoplasm Recurrence, Local/metabolism , Neoplasm Recurrence, Local/mortality , Neoplasm Recurrence, Local/pathology , Neoplasm Recurrence, Local/surgery , Neoplasm Staging , Ovarian Neoplasms/metabolism , Ovarian Neoplasms/mortality , Ovarian Neoplasms/surgery , Prognosis , Prospective Studies , ROC Curve , Survival Rate
17.
Gynecol Oncol ; 131(1): 63-8, 2013 Oct.
Article in English | MEDLINE | ID: mdl-23917082

ABSTRACT

OBJECTIVE: The ovarian cancer-associated ascites is an ideal material for evaluating the interaction between the host immune system and cancer cells in the tumor micro-environment. The aim of this study was to investigate whether the selected target cytokine expression levels in ascites could serve as an immune biomarker for predicting outcomes in ovarian cancer. METHODS: Eighty-eight specimens of ovarian cancer-associated ascites were evaluated to select the target cytokine by a cytokine profiling kit. The 144 total samples were subsequently analyzed for this target cytokine. The correlation between the target cytokine and clinical characteristics was analyzed. RESULTS: Interferon-gamma (IFN-γ) was identified as the target cytokine. Higher levels of IFN-γ in the ascites of the tumor micro-environment were associated with advanced disease (p=0.012), higher tumor histological grading (p=0.004), and sub-optimal surgical status (p=0.040). By multivariate analysis, the adjusted hazard ratios (HRs) were 2.74 (95% confidence interval (CI) 1.85-4.05, p<0.001) for disease-free survival (DFS) and 1.72 (95% CI 1.01-2.93, p=0.048) for overall survival (OS) for a 10-fold increase in IFN-γ concentration in the ascites. An inverse dose-response relationship between IFN-γ level and survival was also noted (Ptrend<0.001 for DFS and Ptrend<0.042 for OS). CONCLUSIONS: Patients with ovarian cancer and higher IFN-γ expression levels in cancer-associated ascites will have shorter DFS and OS. IFN-γ levels in the ascites may be a prognostic marker and a potential reference for immunotherapy targeting IFN-γ.


Subject(s)
Ascites/metabolism , Biomarkers, Tumor/metabolism , Carcinoma/immunology , Carcinoma/pathology , Interferon-gamma/metabolism , Ovarian Neoplasms/immunology , Ovarian Neoplasms/pathology , Adult , Aged , Ascites/etiology , Carcinoma/complications , Disease-Free Survival , Female , Humans , Middle Aged , Neoplasm Grading , Ovarian Neoplasms/complications , Proportional Hazards Models
18.
PLoS One ; 7(10): e47190, 2012.
Article in English | MEDLINE | ID: mdl-23082146

ABSTRACT

BACKGROUND: The regulatory T cells (Tregs) can actively suppress the immune responses. However, literature about detailed changes of host effective and suppressive immunities before and after depletion of Tregs in ovarian carcinomas, is rare. MATERIALS AND METHODS: Ovarian cancer patients and the ascitogenic animal model were employed. Immunologic profiles with flow cytometric analyses, immunohistochemistric staining, RT-PCR, ELISA, and ELISPOT assays were performed. In vivo depletion of Treg cells with the mAb PC61was also performed in the animal model. RESULTS: The cytokines, including IL-4 (p=0.017) and TNF-α (p=0.046), significantly decreased while others such as TGF-ß (p=0.013), IL-6 (p=0.016), and IL-10 (p=0.018) were elevated in ascites of ovarian cancer patients, when the disease progressed to advanced stages. The ratio of CD8(+) T cell/Treg cell in ascites was also lower in advanced diseases than in early diseases (advanced 7.37 ± 0.64 vs. early 14.25 ± 3.11, p=0.037). The kinetic low-dose CD25 Ab depletion group had significantly lower intra-peritoneal tumor weight (0.20 ± 0.03 g) than the sequential high-dose (0.69 ± 0.06 g) and sequential low-dose (0.67 ± 0.07 g) CD25 Ab deletion groups (p=0.001) after 49 days of tumor challenge in the animal. The kinetic low-dose CD25 Ab depletion group generated the highest number of IFN-γ-secreting, mesothelin-specific T lymphocytes compared to the other groups (p<0.001). CONCLUSIONS: The imbalance between effective and suppressive immunities becomes more severe as a tumor progresses. The depletion of Treg cells can correct the imbalance of immunologic profiles and generate potent anti-tumor effects. Targeting Treg cells can be a new strategy for the immunotherapy of ovarian carcinoma.


Subject(s)
Antigens, Neoplasm/immunology , Immunity/immunology , Lymphocyte Depletion , Ovarian Neoplasms/immunology , Ovarian Neoplasms/pathology , T-Lymphocytes, Regulatory/immunology , Animals , Antibodies, Monoclonal/administration & dosage , Antibodies, Monoclonal/immunology , Antibodies, Neoplasm/immunology , Ascites/immunology , CD8-Positive T-Lymphocytes/immunology , Cell Line, Tumor , Cytokines/metabolism , Disease Progression , Dose-Response Relationship, Immunologic , Female , Humans , Interleukin-2 Receptor alpha Subunit/immunology , Kinetics , Lymphocyte Activation/immunology , Mesothelin , Mice , Mice, Inbred C57BL , Neoplasm Staging , Spleen/immunology , Spleen/pathology , Tumor Microenvironment/immunology
19.
Mol Oncol ; 6(3): 360-9, 2012 Jun.
Article in English | MEDLINE | ID: mdl-22265591

ABSTRACT

The alpha-folate receptor (α-FR) is highly-expressed in various non-mucinous tumors of epithelial origin, including ovarian carcinoma. The aim of this study was to investigate the relationship between alpha-folate receptor (α-FR) and the clinico-pathologic features and outcomes of serous ovarian carcinoma patients and the possible mechanism of α-FR to chemo-resistance. Therefore, semi-quantitative reverse-transcription polymerase chain reactions for α-FR expression were performed in the 91 specimens of serous ovarian carcinomas. The expression of α-FR in each ovarian cancer tissue specimen was defined as the ratio of density of α-FR to density of glyceraldehyde-3-phosphate dehydrogenase (GAPDH). In vitro apoptotic experiments were tested in the original OVCAR-3 tumor cells and various OVCAR-3 α-FR-transfectants. Patients with an increased α-FR expression level had poorer responses to chemotherapy (per α-FR expression level increase: odds ratio (OR): 8.97 (95% confidence interval (CI): 1.40-57.36), p = 0.021). An increased α-FR expression level was an independently poor prognostic factor for disease free interval (DFI) (per α-FR expression level increase: hazard ratio (HR): 2.45 (95% CI: 1.16-5.18), p = 0.02) and had a negative impact on overall survival (OS) of these serous ovarian cancer patients (per α-FR expression level increase: HR: 3.6 (95% CI: 0.93-13.29), p = 0.03) by multivariate analyses. α-FR inhibited cytotoxic drug-induced apoptosis in our in vitro apoptotic assays. α-FR could induce chemo-resistance via regulating the expression of apoptosis-related molecules, Bcl-2 and Bax. Therefore, α-FR can be a potential biomarker for the prediction of chemotherapeutic responses and clinical prognosis. It also could be the target of ovarian cancer treatment.


Subject(s)
Cystadenocarcinoma, Serous/drug therapy , Cystadenocarcinoma, Serous/metabolism , Folate Receptor 1/metabolism , Ovarian Neoplasms/metabolism , Antineoplastic Agents/pharmacology , Cell Line, Tumor , Cell Survival/drug effects , Female , Flow Cytometry , Humans , Immunoblotting , Immunoprecipitation , In Vitro Techniques
20.
Biochem J ; 442(2): 293-302, 2012 Mar 01.
Article in English | MEDLINE | ID: mdl-21999204

ABSTRACT

Ovarian cancer has one of the highest mortalities in malignancies in women, but little is known of its tumour progression properties and there is still no effective molecule that can monitor its growth or therapeutic responses. MSLN (mesothelin), a secreted protein that is overexpressed in ovarian cancer tissues with a poor clinical outcome, has been previously identified to activate PI3K (phosphoinositide 3-kinase)/Akt signalling and inhibit paclitaxel-induced apoptosis. The present study investigates the correlation between MSLN and MMP (matrix metalloproteinase)-7 in the progression of ovarian cancer, and the mechanism of MSLN in enhancing ovarian cancer invasion. The expression of MSLN correlated well with MMP-7 expression in human ovarian cancer tissues. Overexpressing MSLN or ovarian cancer cells treated with MSLN showed enhanced migration and invasion of cancer cells through the induction of MMP-7. MSLN regulated the expression of MMP-7 through the ERK (extracellular-signal-regulated kinase) 1/2, Akt and JNK (c-Jun N-terminal kinase) pathways. The expression of MMP-7 and the migrating ability of MSLN-treated ovarian cancer cells were suppressed by ERK1/2- or JNK-specific inhibitors, or a decoy AP-1 (activator protein 1) oligonucleotide in in vitro experiments, whereas in vivo animal experiments also demonstrated that mice treated with MAPK (mitogen-activated protein kinase)/ERK- or JNK-specific inhibitors could decrease intratumour MMP-7 expression, delay tumour growth and extend the survival of the mice. In conclusion, MSLN enhances ovarian cancer invasion by MMP-7 expression through the MAPK/ERK and JNK signal transduction pathways. Blocking the MSLN-related pathway could be a potential strategy for inhibiting the growth of ovarian cancer.


Subject(s)
GPI-Linked Proteins/metabolism , Matrix Metalloproteinase 7/biosynthesis , Ovarian Neoplasms/metabolism , Animals , Base Sequence , Cell Line, Tumor , Cell Movement/genetics , Cell Movement/physiology , DNA Primers/genetics , Female , GPI-Linked Proteins/antagonists & inhibitors , GPI-Linked Proteins/genetics , Gene Expression , Humans , MAP Kinase Signaling System , Matrix Metalloproteinase 7/genetics , Matrix Metalloproteinase Inhibitors , Mesothelin , Mice , Mice, Inbred C57BL , Neoplasm Invasiveness/genetics , Neoplasm Invasiveness/physiopathology , Ovarian Neoplasms/genetics , Ovarian Neoplasms/pathology , RNA, Small Interfering/genetics , Transcription Factor AP-1/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...