Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 29
Filter
1.
Cell Immunol ; 310: 199-204, 2016 12.
Article in English | MEDLINE | ID: mdl-27522179

ABSTRACT

We investigated whether hypoxia-induced activation of Hh signaling contributes to PDL-1 expression in cancer and whether it affects the anti-tumor function of activated lymphocytes. Hypoxia augmented PDL-1 expression and inhibition of Hh signaling reduced PDL-1 expression under hypoxia. When activated lymphocytes were cocultured with cancers treated with a Hh inhibitor, activated lymphocyte cell numbers increased under hypoxia. In contrast, this increase was abrogated when cancer cells were treated with a PDL-1 neutralizing antibody. These results suggest that Hh signaling is one of regulatory pathways of PDL-1 expression under hypoxia and that inhibiting Hh signaling may induce lymphocyte anti-tumor activity.


Subject(s)
B7-H1 Antigen/metabolism , Hedgehog Proteins/antagonists & inhibitors , Hypoxia/immunology , Lymphocytes/immunology , Neoplasms/immunology , Antigens, Neoplasm/immunology , B7-H1 Antigen/genetics , Cell Line, Tumor , Coculture Techniques , DNA-Binding Proteins/genetics , DNA-Binding Proteins/metabolism , Gene Expression Regulation, Neoplastic , Humans , Hypoxia/therapy , Lymphocyte Activation , Molecular Targeted Therapy , Neoplasms/therapy , Nuclear Proteins/genetics , Nuclear Proteins/metabolism , RNA, Small Interfering/genetics , Signal Transduction/drug effects , Signal Transduction/genetics , Smoothened Receptor/genetics , Smoothened Receptor/metabolism , Trans-Activators , Transcription Factors/genetics , Transcription Factors/metabolism , Veratrum Alkaloids/pharmacology , Zinc Finger Protein GLI1/genetics , Zinc Finger Protein GLI1/metabolism
2.
Oncol Rep ; 47(5)2022 May.
Article in English | MEDLINE | ID: mdl-35315500

ABSTRACT

Recently, the cancer microenvironment (CME) has received significant attention. At the local site of the tumor, cancer progression is affected by secreted cytokines and conditions derived from the CME and stimulation by cancer­induced cytokines in an autocrine manner. The CME is characterized by various types of conditions, such as hypoxia, inflammation stimulation, and angiogenesis, and contains various components, such as reactive oxygen species, cancer­associated fibroblasts, infiltrated immune cells, exosomes, and cancer stem cells (CSCs). These conditions and components complicate the progression of cancer. The Hedgehog (HH) signaling pathway is a morphogenesis signaling pathway that is reactivated in some cancers. In these cancers, reactivated HH signaling is involved in the induction of the malignant phenotype. HH signaling is also activated under hypoxic conditions and is considered to be strongly correlated with the CME, including the induction of cancer fibrosis and maintenance of CSCs. The aim of the present review was to elucidate a cancer therapy that targets HH signaling by considering the CME, particularly focusing on hypoxia.


Subject(s)
Hedgehog Proteins , Neoplasms , Hedgehog Proteins/metabolism , Humans , Molecular Targeted Therapy , Neoplasms/pathology , Signal Transduction/genetics , Tumor Microenvironment
3.
Anticancer Res ; 42(6): 2869-2874, 2022 Jun.
Article in English | MEDLINE | ID: mdl-35641270

ABSTRACT

BACKGROUND/AIM: Recently, protein tyrosine phosphatase non-receptor type 3 (PTPN3) has gained attention. However, the role of PTPN3 in cancer has not been fully elucidated. In the present study, we analyzed the role of PTPN3 in pancreatic cancer and investigated whether PTPN3 could be a new therapeutic target for pancreatic cancer. MATERIALS AND METHODS: Two pancreatic ductal adenocarcinoma (PDAC) cell lines were used as target cells. Cell proliferation was investigated using cell counting and a xenograft mouse model. Migration and invasion were analyzed using Transwell inserts. Activation-related signaling molecules were examined by western blotting. RESULTS: PTPN3 contributes to the proliferation, migration, and invasion of PDAC cells in vitro. PTPN3 promotes tumor growth in a mouse xenograft model, an action mediated partially through the MAPK pathway. CONCLUSION: PTPN3 could be a new therapeutic target for pancreatic cancer.


Subject(s)
Carcinoma, Pancreatic Ductal , Pancreatic Neoplasms , Protein Tyrosine Phosphatase, Non-Receptor Type 3 , Animals , Carcinoma, Pancreatic Ductal/genetics , Carcinoma, Pancreatic Ductal/metabolism , Humans , Mice , Pancreatic Neoplasms/drug therapy , Pancreatic Neoplasms/genetics , Pancreatic Neoplasms/metabolism , Protein Tyrosine Phosphatase, Non-Receptor Type 3/metabolism , Pancreatic Neoplasms
4.
Eur J Immunol ; 40(1): 197-203, 2010 Jan.
Article in English | MEDLINE | ID: mdl-19902430

ABSTRACT

CD25+ FOXP3+CD4+ T cells (Treg) have been considered to play an important role in immune tolerance against several tumor antigens. It has also been indicated that high-level expression of FOXP3 (FOXP3high) is sufficient to confer suppressive activity to normal non-Treg. Here, we showed for the first time that vascular endothelial growth factor receptor 2 (VEGFR2) is selectively expressed by FOXP3high but not FOXP3low Treg. Such VEGFR2+ Treg exist in several tissues including PBMC and malignant effusion-derived lymphocytes. In conclusion, VEGFR2 may be a novel target for controlling Treg with highly suppressive function.


Subject(s)
Forkhead Transcription Factors/immunology , T-Lymphocytes, Regulatory/immunology , Vascular Endothelial Growth Factor Receptor-2/immunology , Humans , Immunotherapy , Neoplasms/immunology , Neoplasms/therapy , Transforming Growth Factor beta/immunology
5.
Anticancer Res ; 41(8): 4047-4052, 2021 Aug.
Article in English | MEDLINE | ID: mdl-34281873

ABSTRACT

BACKGROUND/AIM: Tropomyosin-related kinase B (TrkB)/brain-derived neurotrophic factor (BDNF) signaling plays a role in inducing malignant phenotypes in several aggressive types of cancers. To create a conclusive therapy targeting TrkB/BDNF signaling in solid refractory cancers, the biological significance of TrkB/BDNF signaling was analyzed in pancreatic ductal adenocarcinoma (PDAC) cells. MATERIALS AND METHODS: Three PDAC cell lines were used as target cells to investigate proliferation and invasiveness. Small interfering RNA (siRNA) and the TrkB tyrosine kinase inhibitor k252a were used as TrkB/BDNF signaling inhibitors. RESULTS: All PDAC cell lines expressed TrkB and BDNF. When TrkB and BDNF were inhibited by siRNA or k252a, the invasiveness of PANC-1 and SUIT-2 cells significantly decreased. When TrkB was inhibited by siRNA or k252a, proliferation was significantly inhibited in PDAC cells. CONCLUSION: TrkB/BDNF signaling may be a new therapeutic target for PDAC. Therapies targeting TrkB/BDNF signaling may be a conclusive cancer therapy for refractory solid cancer.


Subject(s)
Brain-Derived Neurotrophic Factor/metabolism , Carcinoma, Pancreatic Ductal/metabolism , Membrane Glycoproteins/metabolism , Pancreatic Neoplasms/metabolism , Receptor, trkB/metabolism , Brain-Derived Neurotrophic Factor/genetics , Carbazoles/pharmacology , Carcinoma, Pancreatic Ductal/pathology , Cell Line, Tumor , Cell Proliferation/drug effects , Humans , Indole Alkaloids/pharmacology , Membrane Glycoproteins/antagonists & inhibitors , Membrane Glycoproteins/genetics , Pancreatic Neoplasms/pathology , Protein Kinase Inhibitors/pharmacology , RNA Interference , RNA, Small Interfering/genetics , Receptor, trkB/antagonists & inhibitors , Receptor, trkB/genetics , Signal Transduction/drug effects
6.
Cancer Immunol Immunother ; 59(5): 653-61, 2010 May.
Article in English | MEDLINE | ID: mdl-19908042

ABSTRACT

The human immune system consists of a balance between immune surveillance against non-self antigens and tolerance of self-antigens. CD8(+) T cells and CD4(+) regulatory T cells (Tregs) are the main players for immune surveillance and tolerance, respectively. We examined immunohistochemically the immunological balance at the tumor site using 94 surgically resected colorectal cancer tissues. Forkhead box P3 (FOXP3)(+) cells were considered to be Tregs in the present study. The number of intratumoral FOXP3(+) cells (itFOXP3(+) cells) was positively correlated with lymph node metastases (P = 0.030). itCD8(+) T/itFOXP3(+) cell ratio negatively correlated with pathological stages (P = 0.048). Next, relationship between the number of itCD8(+) T cells or itFOXP3(+) cells and survival prognosis in 94 patients who underwent a curative resection was analyzed. Only itCD8(+) T/itFOXP3(+) cell ratio positively correlated with disease-free survival (0.023) and overall survival (P = 0.010). Multivariate analysis indicated that itCD8(+) T/itFOXP3(+) cell ratio is an independent prognostic factor (P = 0.035) of overall survival. The number of itFOXP3(+) cells positively correlated with transforming growth factor-beta TGF-beta production at the tumor site (P = 0.020). In conclusion, itCD8(+) T/itFOXP3(+) cell ratio is a predictive marker for both disease-free survival time and overall survival time in patients with colorectal cancer. Importantly, itCD8(+) T/itFOXP3(+) cell ratio may be an independent prognostic factor. And, tumor-producing TGF-beta may contribute to the increased number of itFOXP3(+) cells.


Subject(s)
Biomarkers, Tumor/immunology , CD8-Positive T-Lymphocytes/immunology , Colorectal Neoplasms/immunology , Lymphocytes, Tumor-Infiltrating/immunology , T-Lymphocyte Subsets/immunology , T-Lymphocytes, Regulatory/immunology , Aged , Biomarkers, Tumor/metabolism , CD8-Positive T-Lymphocytes/metabolism , Cell Separation , Colorectal Neoplasms/metabolism , Colorectal Neoplasms/mortality , Disease-Free Survival , Female , Flow Cytometry , Forkhead Transcription Factors/immunology , Forkhead Transcription Factors/metabolism , Humans , Immunohistochemistry , Kaplan-Meier Estimate , Lymphatic Metastasis/immunology , Lymphatic Metastasis/pathology , Lymphocytes, Tumor-Infiltrating/metabolism , Male , Middle Aged , Prognosis , T-Lymphocyte Subsets/metabolism , T-Lymphocytes, Regulatory/metabolism , Transforming Growth Factor beta/metabolism
7.
Cancer Immunol Immunother ; 59(5): 675-86, 2010 May.
Article in English | MEDLINE | ID: mdl-19862523

ABSTRACT

Recently, it was reported that Hh signaling is activated in tumor stromal cells but not in tumor cells themselves and that stromal cells may play a role in the proliferation of cancer cells. This suggests the possibility that stromal cells have an important role in the proliferation of tumor cells that may be mediated through Hh signaling. In this report, we present for the first time that inflammation-stimulated monocytes produce Shh through activation of the NF-kappaB signaling pathway, and that the Shh produced promotes the proliferation of pancreatic cancer cells in a paracrine manner through Hh signaling.


Subject(s)
Carcinoma, Pancreatic Ductal/metabolism , Gene Expression Regulation, Neoplastic , Hedgehog Proteins/biosynthesis , Monocytes/metabolism , NF-kappa B/metabolism , Pancreatic Neoplasms/metabolism , Cell Line, Tumor , Cell Proliferation , Electrophoretic Mobility Shift Assay , Fluorescent Antibody Technique , Gene Expression , Humans , Immunoblotting , Immunohistochemistry , RNA, Small Interfering , Reverse Transcriptase Polymerase Chain Reaction , Signal Transduction/physiology , Transduction, Genetic
8.
Cancer Lett ; 484: 9-15, 2020 08 01.
Article in English | MEDLINE | ID: mdl-32380129

ABSTRACT

Chemotherapy and immunotherapy for pancreatic ductal adenocarcinoma (PDAC) have limited success. One reason for this is thought to be the cancer microenvironment surrounding PDAC. Hypoxia is a feature of the cancer microenvironment. Under hypoxia, different various molecules and signaling pathways are activated compared with normoxia. To develop a new effective therapeutic strategy for PDAC, we need to target these hypoxic conditions to overcome PDAC. To inhibit the malignant phenotype, the cellular changes that occur under hypoxia should be elucidated. Various molecules and signaling that are activated by hypoxia may contribute to the induction of malignant phenotypes of PDAC such as proliferation, invasion, tumorigenesis, chemosensitivity, and autophagy. If we can develop therapeutic approaches to target one of these molecules or signaling pathways, we may proceed to the next therapeutic step of successfully treating refractory PDAC.


Subject(s)
Carcinoma, Pancreatic Ductal/genetics , Gene Expression Regulation, Neoplastic/genetics , Hypoxia , Pancreatic Neoplasms/genetics , Tumor Microenvironment/genetics , Antineoplastic Agents/pharmacology , Carcinoma, Pancreatic Ductal/metabolism , Carcinoma, Pancreatic Ductal/therapy , Cell Line, Tumor , Gene Expression Regulation, Neoplastic/drug effects , Humans , Hypoxia-Inducible Factor 1, alpha Subunit/genetics , Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , Immunotherapy/methods , Pancreatic Neoplasms/metabolism , Pancreatic Neoplasms/therapy , Tumor Microenvironment/drug effects
9.
J Surg Oncol ; 100(8): 725-31, 2009 Dec 15.
Article in English | MEDLINE | ID: mdl-19722233

ABSTRACT

BACKGROUND: Inflammation plays a multifaceted role in cancer progression, and NF-kappaB is one of the key factors connecting inflammation with cancer progression. We have shown that lipopolysaccharide (LPS) promotes NF-kappaB activation in colon cancer cells and pancreatic cancer cells. However, it is unclear why inflammatory stimuli can induce NF-kappaB activation in cancer cells. METHODS: We used two human pancreatic cancer cells, Panc-1 and AsPC-1, as target cells. LPS was used as an inflammatory stimulus. To confirm the participation of TLR4/NF-kappaB signaling pathway, we used three different NF-kappaB inhibitors (PDTC, IkappaBalpha mutant, and NF-kappaB decoy ODN) and siRNAs (against TLR4, MyD88, and MMP-9). Effect of LPS on pancreatic cancer cell invasive ability was determined by Matrigel invasion assay. RESULTS: LPS increased the invasive ability of pancreatic cancer cells, while blockade of NF-kappaB pathway decreased the LPS-dependent increased invasive ability. Blockade of TLR4 or MyD88 by siRNA also decreased the LPS-dependent increased invasive ability. CONCLUSION: These results suggest that TLR/MyD88/NF-kappaB signaling pathway plays a significant role in connecting inflammation and cancer invasion and progression.


Subject(s)
Lipopolysaccharides/toxicity , Myeloid Differentiation Factor 88/physiology , Pancreatic Neoplasms/pathology , Signal Transduction/physiology , Toll-Like Receptor 4/physiology , Cell Line, Tumor , Humans , Myeloid Differentiation Factor 88/analysis , NF-kappa B/antagonists & inhibitors , NF-kappa B/metabolism , Neoplasm Invasiveness , Toll-Like Receptor 4/analysis
10.
Anticancer Res ; 29(3): 871-9, 2009 Mar.
Article in English | MEDLINE | ID: mdl-19414322

ABSTRACT

Understanding the expression patterns of estrogen receptor-alpha (ERalpha) is essential for determining therapeutic strategies for patients with breast cancer. The prognosis of patients with ERalpha-negative breast cancer is still poor. We have previously shown that Hedgehog (Hh) signaling is constitutively activated in breast cancer and that Hh signaling could be a new therapeutic target. Therefore, in this study, whether or not Hh signaling could be utilized as a therapeutic target for patients with ERalpha-negative breast cancer was examined. For this purpose, three ERalpha-negative breast cancer cell lines were used in which Hh pathway-related molecules such as the ligand Patched1 and the transcriptional factor Gli1 as target cells are expressed. Cyclopamine, an inhibitor of the Hh pathway, significantly suppressed both the cell proliferation and invasion ability of these cancer cells. In addition, the knockdown of Gli1 by RNA interference in these cells also significantly reduced both cell proliferation and invasion ability. Since our previous data have shown a constitutive activation of the Hh pathway in surgically-resected ERalpha-negative breast cancer specimens, the Hh pathway, especially Gli1, may be a useful therapeutic target for patients with ERalpha-negative breast cancer.


Subject(s)
Adenocarcinoma/metabolism , Breast Neoplasms/metabolism , Carcinoma, Papillary/metabolism , Estrogen Receptor alpha/metabolism , Hedgehog Proteins/metabolism , Transcription Factors/metabolism , Adenocarcinoma/genetics , Adenocarcinoma/secondary , Aged , Breast Neoplasms/genetics , Breast Neoplasms/pathology , Carcinoma, Papillary/genetics , Carcinoma, Papillary/secondary , Cell Proliferation/drug effects , Female , Hedgehog Proteins/antagonists & inhibitors , Hedgehog Proteins/genetics , Humans , Immunoenzyme Techniques , Middle Aged , Neoplasm Invasiveness , Prognosis , RNA, Messenger/genetics , RNA, Messenger/metabolism , RNA, Small Interfering/pharmacology , Reverse Transcriptase Polymerase Chain Reaction , Signal Transduction , Transcription Factors/antagonists & inhibitors , Transcription Factors/genetics , Tumor Cells, Cultured , Veratrum Alkaloids/pharmacology , Zinc Finger Protein GLI1
11.
Anticancer Res ; 29(3): 881-8, 2009 Mar.
Article in English | MEDLINE | ID: mdl-19414323

ABSTRACT

It has been suggested that immunosuppressive cytokines such as transforming growth factor beta (TGF-beta) and interleukin 10 play an important role in the induction and/or maintenance of regulatory T-cells (Tregs) in patients with cancer. In the present study, whether or not vascular endothelial growth factor (VEGF) contributes to the induction and/or maintenance of Tregs was examined, because of experience with a patient in whom a positive correlation between VEGF concentration and the percentage of Tregs (% Tregs) among the total CD4(+) T-cells in the pleural effusion was found during dendritic cell activated lymphocyte therapy. CD4(+)CD25(high) T-cells were estimated as Tregs in the present study. In an in vitro experimental system, VEGF-containing malignant effusions increased the % Tregs in autologous peripheral blood mononuclear cells (PBMCs), which could be suppressed by the addition of a humanized monoclonal anti-VEGF antibody (bevacizumab [Avastin]). When VEGF-producing hepatic carcinoma cells were mix-cultured with PBMCs, the % Tregs increased and this increase was also suppressed by the addition of bevacizumab. Whether or not bevacizumab can affect the % Tregs of PBMCs in patients with colon cancer was also examined. Three out of four patients showed a significant decrease of the % Tregs after intravenous injection of bevacizumab. Interestingly, the expression of VEGF receptor-2 (VEGFR-2) was higher in Tregs than in other CD4(+) T-cells. Taken together, the data presented here indicate a contribution of VEGF to induction and/or maintenance of Tregs in patients with cancer.


Subject(s)
Neoplasms/immunology , Pleural Effusion, Malignant/immunology , T-Lymphocytes, Regulatory/immunology , Vascular Endothelial Growth Factor A/physiology , Adult , Aged , Angiogenesis Inhibitors/therapeutic use , Antibodies, Monoclonal/therapeutic use , Antibodies, Monoclonal, Humanized , Bevacizumab , Dendritic Cells/transplantation , Enzyme-Linked Immunosorbent Assay , Female , Flow Cytometry , Humans , Immunotherapy , Male , Middle Aged , Neoplasms/therapy , Pleural Effusion, Malignant/therapy , Vaccination , Vascular Endothelial Growth Factor A/antagonists & inhibitors , Vascular Endothelial Growth Factor Receptor-2/metabolism
12.
Anticancer Res ; 29(3): 843-50, 2009 Mar.
Article in English | MEDLINE | ID: mdl-19414318

ABSTRACT

BACKGROUND: We have reported previously that docetaxel (TXT) induces apoptosis and nuclear factor-kappaB (NF-kappaB) activation, and that blockade of NF-kappaB activation augments TXT-induced apoptosis in human gastric cancer cells. In addition, we have also shown that a protein-bound polysaccharide PSK enhances TXT-induced apoptosis through NF-kappaB inhibition in human pancreatic cancer cells. Based on these observations, in the present study the possibility that PSK could enhance TXT-mediated tumor suppression was examined in vivo and in vitro. MATERIALS AND METHODS: A gastric cancer xenograft model was used to examine the enhanced TXT-mediated tumor suppression by PSK in vivo. The effects of PSK on proliferation and apoptosis induced by TXT in gastric cancer cells were evaluated in vitro using a human gastric cancer cell line, MK-1. The effect of PSK on increased TXT-induced invasion was also measured. RESULTS: PSK enhanced TXT-mediated tumor suppression in vivo. Immunohistochemical analyses of the tumors revealed that TXT increased NF-kappaB activation in the tumors and this was suppressed by PSK. In the ex vivo experimental system, PSK enhanced the growth inhibition and apoptosis induced by TXT in the MK-1 cells and reduced the increased invasive ability induced by TXT. CONCLUSION: PSK enhanced TXT-induced tumor suppression in a gastric cancer xenograft model.


Subject(s)
Adenocarcinoma/drug therapy , Antineoplastic Agents/therapeutic use , Proteoglycans/administration & dosage , Stomach Neoplasms/drug therapy , Taxoids/administration & dosage , Adenocarcinoma/secondary , Animals , Apoptosis/drug effects , Cell Movement/drug effects , Cell Proliferation/drug effects , Docetaxel , Drug Synergism , Electrophoretic Mobility Shift Assay , Female , Humans , Immunoenzyme Techniques , Mice , Mice, Inbred BALB C , Mice, Nude , NF-kappa B/genetics , NF-kappa B/metabolism , Neoplasm Invasiveness , RNA, Messenger/genetics , RNA, Messenger/metabolism , Reverse Transcriptase Polymerase Chain Reaction , Stomach Neoplasms/pathology , Survival Rate , Tumor Cells, Cultured , Xenograft Model Antitumor Assays
13.
Anticancer Res ; 39(3): 1179-1184, 2019 Mar.
Article in English | MEDLINE | ID: mdl-30842147

ABSTRACT

BACKGROUND/AIM: Small-cell lung cancer (SCLC) remains one of deadliest types of cancers. Cis-diamminedichloroplatinum (CDDP) is a key chemotherapeutic agent for SCLC, however, its therapeutic effect is limited. Recently, hypoxia in the cancer microenvironment has been suggested to influence the effect of cancer therapy. MATERIALS AND METHODS: Using small interfering RNA inhibition of leukocyte common antigen-related interacting protein alpha 4 (liprin-α4), and of hypoxia-inducible factor (HIF)-1α, proliferation, invasion, migration and chemosensitivity were investigated in SBC-5 SCLC cells, under normoxia and hypoxia. RESULTS: Liprin-α4 was found to contribute to proliferation, but not migration and invasion of SBC-5 cells both under normoxia and hypoxia. Inhibition of liprin-α4 increased chemosensitivity of SBC-5 cells under hypoxia. Liprin-α4 signaling occurs through mitogen-activated protein kinase pathways via activation of HIF1α expression. Inhibition of HIF1α reduced proliferation and increased chemosensitivity of SBC-5 cells under hypoxia. CONCLUSION: Liprin-α4 inhibition may enhance the effect of CDDP and liprin-α4 might be a novel therapeutic target in SCLC.


Subject(s)
Hypoxia-Inducible Factor 1, alpha Subunit/genetics , Lung Neoplasms/therapy , RNA, Small Interfering/administration & dosage , Receptor-Like Protein Tyrosine Phosphatases/genetics , Small Cell Lung Carcinoma/therapy , Antineoplastic Agents/pharmacology , Cell Line, Tumor , Cisplatin/pharmacology , Humans , Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , Lung Neoplasms/genetics , Lung Neoplasms/metabolism , Receptor-Like Protein Tyrosine Phosphatases/metabolism , Small Cell Lung Carcinoma/genetics , Small Cell Lung Carcinoma/metabolism
14.
Anticancer Res ; 28(4C): 2401-8, 2008.
Article in English | MEDLINE | ID: mdl-18751426

ABSTRACT

We previously showed that a combination therapy with tumor cell-pulsed monocyte-derived dendritic cells (DCs) and activated lymphocytes was well tolerated in patients with disseminated carcinomas. Recently, accumulating evidence has indicated that regulatory T-cells (Tregs), a unique population of CD4+ T-cells, are increased in patients with several advanced malignancies and prevent cell-mediated immune responses against tumors. However, reports analyzing the relationship between the Tregs population and the effects of immunotherapy are extremely rare. In the present study, 22 patients received an intravenous injection of DC-activated lymphocytes (DAK) and/or a subcutaneous injection of tumor-pulsed DCs (DC vaccine) every 2 to 4 weeks. The Tregs were defined based on their expression of CD4, CD25 and FOXP3, a transcription factor. Most CD4+CD25high T-cells expressed FOXP3. Therefore, CD4+CD25high T-cells were evaluated as Tregs in the present study. As reported previously, the percentage of Tregs (% Tregs) among total CD4+ T-cells in peripheral blood mononuclear cells (PBMCs) was significantly higher for advanced cancer patients than for healthy volunteers. When the patients were divided into three groups according to their survival time, i.e. 12 short-survival patients, 4 medium-survival patients and 6 long-survival patients, the % Tregs of the long-survival patients before the therapy was significantly lower than that of the short-survival patients (p=0.026). The % Tregs decreased after the therapy, although the difference did not reach statistical significance. When the patients were divided into a high group (>4.99%: 7 patients) and a low group (<4.99%: 15 patients) according to their % Tregs before the therapy, the survival times of the two groups differed significantly (p=0.0034). These data suggest that the % Tregs among the PBMCs might be used as an effect prediction factor of immunotherapy for patients with advanced cancer.


Subject(s)
Immunotherapy, Adoptive , Neoplasms/immunology , Neoplasms/therapy , T-Lymphocytes, Regulatory/immunology , Adult , Aged , Dendritic Cells/immunology , Female , Flow Cytometry , Humans , Leukocytes, Mononuclear/immunology , Male , Middle Aged , Predictive Value of Tests , Prognosis
15.
Hepatogastroenterology ; 55(84): 859-60, 2008.
Article in English | MEDLINE | ID: mdl-18705283

ABSTRACT

A 53-year-old Japanese female was referred to Miyazaki University hospital for the surgical treatment of gastric cancer. She had no symptoms and the presence of the complex cystic gallbladder with multiple and linear hyperechogenic septa was incidentally found by ultrasonography. Multicystic lesion of gallbladder was also observed on DIC-CT and MRCP. Neither the bile duct nor the pancreatic duct was dilated. The papilla of the Vater was of normal appearance endoscopically. Pathological study of the biopsy specimen from the type 3 tumor of the stomach showed poorly differentiated adenocarcinoma. Distal partial gastrectomy and cholecystectomy were performed. The gross examination of the gallbladder confirmed the presence of multiple thin septa with honeycomb appearance. Cancer cells were not observed histologically and final diagnosis was the multiseptate gallbladder. The postoperative course was uneventful. We herein report a rare case of multiseptate gallbladder showing the findings on DIC-CT and MRCP.


Subject(s)
Cholangiography , Cholangiopancreatography, Magnetic Resonance , Gallbladder/abnormalities , Image Processing, Computer-Assisted , Imaging, Three-Dimensional , Tomography, X-Ray Computed , Ultrasonography , Adenocarcinoma/diagnosis , Adenocarcinoma/pathology , Adenocarcinoma/surgery , Cholecystectomy , Female , Gallbladder/pathology , Humans , Incidental Findings , Middle Aged , Stomach Neoplasms/diagnosis , Stomach Neoplasms/pathology , Stomach Neoplasms/surgery
16.
Anticancer Res ; 38(4): 1979-1986, 2018 04.
Article in English | MEDLINE | ID: mdl-29599313

ABSTRACT

We previously reported that brain-derived neurotrophic factor (BDNF)/neurotrophic receptor tyrosine kinase 2 (NTRK2/TRKB) signaling contributes to induction of malignant phenotype of gallbladder cancer (GBC). Recently, pan-TRK inhibitors have been evaluated and their dramatic clinical activity is being shown for a variety of cancer types harboring an NTRK rearrangement in phase I trials. ONO-7579 is an oral pan-TRK inhibitor currently under investigation in phase I/II clinical trial for TRK-rearranged solid tumors. In this study, we evaluated the anticancer effect of ONO-7579 using GBC cells with or without KRAS mutant, NOZ, TYGBK-1. Our study showed that ONO-7579 had a suppressive effect on GBC proliferation in TYGBK-1, and on invasive potential and vascular endothelial growth factor expression in TYGBK-1 and NOZ. Our data indicated that ONO-7579 could be a promising treatment option for patients with GBC.


Subject(s)
Gallbladder Neoplasms/drug therapy , Membrane Glycoproteins/antagonists & inhibitors , Organic Chemicals/pharmacology , Protein Kinase Inhibitors/pharmacology , Receptor, trkB/antagonists & inhibitors , Brain-Derived Neurotrophic Factor/antagonists & inhibitors , Cell Line, Tumor , Cell Movement/drug effects , Cell Proliferation/drug effects , Epithelial-Mesenchymal Transition/drug effects , Gallbladder Neoplasms/enzymology , Gallbladder Neoplasms/metabolism , Gallbladder Neoplasms/pathology , Humans , Membrane Glycoproteins/biosynthesis , Neoplasm Invasiveness , Organic Chemicals/chemistry , Receptor, trkB/biosynthesis , Vascular Endothelial Growth Factor A/biosynthesis
17.
Anticancer Res ; 38(8): 4543-4547, 2018 Aug.
Article in English | MEDLINE | ID: mdl-30061220

ABSTRACT

BACKGROUND/AIM: Small cell lung cancer (SCLC) is still a deadly type of cancer for which there are few effective therapeutic strategies. Development of a new molecule targeting agent is urgently desired. Previously we showed that recombination signal binding protein for immunoglobulin-kappa-J region (RBPJ) and mastermind-like 3 (MAML3) are new therapeutic targets for pancreatic cancer. In the present study, we analyzed whether RBPJ/MAML3 inhibition could also be a new therapeutic strategy for SCLC. MATERIALS AND METHODS: Using silencing of RBPJ/MAML3, proliferation, invasion, migration and chemosensitivity of SBC-5 cells were investigated. RESULTS: RBPJ/MAML3 inhibition reduced Smoothened and HES1 expression, suggesting that RBPJ/MAML3 signaling was through Hedgehog and NOTCH pathways. In the analysis of cell functions, RBPJ/MAML3 inhibition significantly reduced proliferation and invasiveness via reduction of expression of matrix metalloproteinases. On the other hand, RBPJ/MAML3 inhibition also reduced chemosensitivity to cis-diamminedichlo-roplatinum and gemcitabine. CONCLUSION: These results suggest that RBPJ and MAML3 could be new therapeutic targets for SCLC, however, chemosensitivity may be reduced in combinational use with other chemo-therapeutic agents.


Subject(s)
DNA-Binding Proteins/metabolism , Immunoglobulin J Recombination Signal Sequence-Binding Protein/metabolism , Lung Neoplasms/metabolism , Nuclear Proteins/metabolism , Small Cell Lung Carcinoma/metabolism , Transcription Factors/metabolism , Antineoplastic Combined Chemotherapy Protocols/pharmacology , Cell Line, Tumor , Cell Movement/drug effects , Cell Proliferation/drug effects , Deoxycytidine/analogs & derivatives , Deoxycytidine/pharmacology , Hedgehog Proteins/metabolism , Humans , Lung Neoplasms/drug therapy , Matrix Metalloproteinases/metabolism , Neoplasm Invasiveness/pathology , Pancreatic Neoplasms/drug therapy , Pancreatic Neoplasms/metabolism , Receptors, Notch/metabolism , Signal Transduction/drug effects , Small Cell Lung Carcinoma/drug therapy , Trans-Activators , Gemcitabine
18.
Anticancer Res ; 37(2): 561-565, 2017 02.
Article in English | MEDLINE | ID: mdl-28179302

ABSTRACT

The role of cluster of differentiation (CD) 24 in breast cancer remains unclear; previously, we showed that CD24 suppresses malignant phenotypes by inactivating Hedgehog signaling through signal transducer and activator of transcription (STAT) 1 inhibition. In this study, we examined how CD24 affects chemosensitivity in breast cancer cells. The CD44+CD24+ breast cancer cell line MCF-7 was transfected with CD24 with/without STAT1 siRNA, and chemosensitivity to 5-fluorouracil (5-FU) and cis-diamminedichloroplatinum (CDDP) was measured. CD24 inhibition reduced chemosensitivity to 5-FU, while STAT1 inhibition did not affect chemosensitivity to 5-FU in CD24 siRNA-transfected cells. Conversely, CD24 inhibition did not affect chemosensitivity to CDDP, while STAT1 inhibition reduced chemosensitivity to CDDP in CD24 siRNA-transfected cells. STAT1 inhibition, but not CD24 inhibition, reduced expression of the ATP-binding cassette (ABC) transporter genes, ABCB1 and ABCG2. In conclusion, CD24 inhibition may modulate chemosensitivity according to drug type, but ABC transporter expression appears not to contribute to this mechanism. This study contributes to determining the role of CD24 in breast cancer.


Subject(s)
Breast Neoplasms/drug therapy , Breast Neoplasms/metabolism , CD24 Antigen/metabolism , ATP Binding Cassette Transporter 1/biosynthesis , ATP Binding Cassette Transporter, Subfamily B/biosynthesis , ATP Binding Cassette Transporter, Subfamily G, Member 2/biosynthesis , Breast Neoplasms/genetics , CD24 Antigen/genetics , Female , Fluorouracil/pharmacology , Humans , MCF-7 Cells , Neoplasm Proteins/biosynthesis , RNA, Small Interfering/administration & dosage , RNA, Small Interfering/genetics , STAT1 Transcription Factor/antagonists & inhibitors , STAT1 Transcription Factor/genetics , STAT1 Transcription Factor/metabolism , Transfection
19.
Anticancer Res ; 37(9): 4987-4992, 2017 09.
Article in English | MEDLINE | ID: mdl-28870922

ABSTRACT

BACKGROUND/AIM: Previously we have shown that tropomyosin-related kinase B (TRKB) and Hedgehog (Hh) signalling pathways induce malignant phenotypes in many cancer types. However, results from small cell lung cancer (SCLC) clinical trials using TRK and Hh inhibitors have been disappointing. One reason for this may be the existence of crosstalk between TRKB and Hh signalling pathways. In this study, we detected negative crosstalk between the TRKB and Hh-GLI1 signalling pathways. MATERIALS AND METHODS: The human small cell lung carcinoma cell line, SBC-5, was used. Using small interfering RNA to inhibit TRKB and Hh signalling, whether TRKB and Hh signaling contribute to proliferation and invasiveness in SBC-5 cells were investigated. RESULTS: TRKB expression in GLI1 siRNA-transfected SBC-5 cells was higher than that of control cells. GLI1-knockdown alone did not affect invasiveness of SBC-5 cells. However, combined knockdown of TRKB and GLI1 significantly decreased invasiveness. Moreover, combined TRKB and GLI1 knockdown inhibited proliferation and migration to a greater extent than when either was inhibited alone. CONCLUSION: These results suggest that Hh inhibition increases TrkB expression to counter tumor suppression in SBC-5 cells. The combined use of TRKB and Hh inhibitors may, therefore, be useful for the treatment of refractory SCLC.


Subject(s)
Cell Movement , Cell Proliferation , Hedgehog Proteins/antagonists & inhibitors , Lung Neoplasms/pathology , Membrane Glycoproteins/antagonists & inhibitors , Receptor, trkB/antagonists & inhibitors , Small Cell Lung Carcinoma/pathology , Zinc Finger Protein GLI1/antagonists & inhibitors , Apoptosis , Hedgehog Proteins/genetics , Humans , Lung Neoplasms/genetics , Lung Neoplasms/metabolism , RNA, Small Interfering/genetics , Small Cell Lung Carcinoma/genetics , Small Cell Lung Carcinoma/metabolism , Tumor Cells, Cultured
20.
Anticancer Res ; 37(12): 6649-6654, 2017 12.
Article in English | MEDLINE | ID: mdl-29187440

ABSTRACT

BACKGROUND/AIM: In pancreatic cancer, where the microenvironment is extremely hypoxic, analyzing signal transduction under hypoxia is thought to be significantly important. By investigating microarray analysis of pancreatic cancer cells cultured under both normoxia and hypoxia, we found that the expression of leukocyte common antigen-related (LAR)-interacting protein (liprin)-α4 was extremely increased under hypoxia compared to under normoxia. MATERIALS AND METHODS: In the present study, the biological significance of liprin-α4 in pancreatic cancer was investigated and whether liprin-α4 has potential as a therapeutic target for pancreatic cancer was estimated. RESULTS: Suppression of liprin-α4 reduced proliferation of pancreatic cancer cells both in vitro and in vivo. Inhibition of liprin-α4 also reduced invasiveness through the suppression of endothelial-mesenchymal transition. Stimulation by liprin-α4 was through phosphoinositide 3-kinase and mitogen-activated protein kinase signaling pathways. CONCLUSION: Liprin-α4 plays a pivotal role in inducing malignant phenotypes such as increased proliferation and invasion in pancreatic cancer, and that liprin-α4 could be a new effective therapeutic target for pancreatic cancer.


Subject(s)
Carcinoma, Pancreatic Ductal/genetics , Gene Expression Profiling/methods , Gene Expression Regulation, Neoplastic , Pancreatic Neoplasms/genetics , Receptor-Like Protein Tyrosine Phosphatases/genetics , Animals , Carcinoma, Pancreatic Ductal/metabolism , Carcinoma, Pancreatic Ductal/pathology , Cell Hypoxia , Cell Line, Tumor , Cell Proliferation/genetics , Female , Humans , Hypoxia , Mice, Inbred BALB C , Mice, Nude , Mitogen-Activated Protein Kinases/metabolism , Pancreatic Neoplasms/metabolism , Pancreatic Neoplasms/pathology , Phosphatidylinositol 3-Kinases/metabolism , RNA Interference , Signal Transduction , Transplantation, Heterologous
SELECTION OF CITATIONS
SEARCH DETAIL