Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 36
Filter
1.
J Immunother ; 47(3): 89-97, 2024 Apr 01.
Article in English | MEDLINE | ID: mdl-38297883

ABSTRACT

In the tumor microenvironment, wherein cytotoxic lymphocytes interact with cancer cells, lymphocyte exhaustion, an immune checkpoint inhibitor target, is promoted. However, the efficacy of these inhibitors is limited, and improving response rates remains challenging. We previously reported that protein tyrosine phosphatase nonreceptor type (PTPN) 3 is a potential immune checkpoint molecule for activated lymphocytes and that PTPN3 inhibition should be a focus area for cancer immunotherapy development. Therefore, in this study, we focused on PTPN3-suppressive therapy in terms of lymphocyte exhaustion under hypoxic conditions, which are a cancer microenvironment, and investigated measures for improving the response to anti-programmed death receptor (PD)-1 antibody drugs. We found that PTPN3 expression was upregulated in activated lymphocytes under hypoxic conditions, similar to the findings for other immune checkpoint molecules, such as PD-1, T cell immunoglobulin mucin-3, and lymphocyte-activation gene-3; furthermore, it functioned as a lymphocyte exhaustion marker. In addition, PTPN3-suppressed activated lymphocytes promoted the mammalian target of rapamycin (mTOR)-Akt signaling pathway activation and enhanced proliferation, migration, and cytotoxic activities under hypoxic conditions. Furthermore, PTPN3 suppression in activated lymphocytes increased PD-1 expression and enhanced the antitumor effects of anti-PD-1 antibody drugs against head and neck cancer in vitro and in vivo. These results suggest that the suppression of PTPN3 expression in activated lymphocytes enhances the therapeutic effect of anti-PD-1 antibody drugs in head and neck cancer, especially under hypoxic conditions that cause lymphocyte exhaustion.


Subject(s)
Antineoplastic Agents , Head and Neck Neoplasms , Humans , Programmed Cell Death 1 Receptor , Head and Neck Neoplasms/drug therapy , Lymphocytes/metabolism , Immunotherapy , Tumor Microenvironment , Protein Tyrosine Phosphatase, Non-Receptor Type 3/metabolism
2.
Front Immunol ; 14: 1223331, 2023.
Article in English | MEDLINE | ID: mdl-37881436

ABSTRACT

Neoantigens/ are tumor-specific antigens that evade central immune tolerance mechanisms in the thymus. Long-term tumor-specific cytotoxic T lymphocyte activity maintenance requires class II antigen-reactive CD4+ T cells. We had previously shown that intranodal vaccination with class I neoantigen peptide-pulsed dendritic cells (DCs) induced a robust immune response in a subset of patients with metastatic cancer. The present study aimed to perform a detailed ex vivo analysis of immune responses in four patients receiving an intranodal hybrid human leukocyte antigen class II neoantigen peptide encompassing a class I neoantigen epitope (hybrid neoantigen)-pulsed DC vaccine. After vaccination, strong T-cell reactions against the hybrid class II peptide and the class I-binding neoantigen peptide were observed in all four patients. We found that hybrid class II neoantigen peptide-pulsed DCs stimulated CD4+ T cells via direct antigen presentation and CD8+ T cells via cross-presentation. Further, we demonstrated that hybrid class II peptides encompassing multiple class I neoantigen epitope-pulsed DCs could present multiple class I peptides to CD8+ T cells via cross-presentation. Our findings provide insight into the mechanisms underlying hybrid neoantigen-pulsed DC vaccine therapy and suggest future neoantigen vaccine design.


Subject(s)
CD8-Positive T-Lymphocytes , Neoplasms , Humans , Antigens, Neoplasm , Peptides , Epitopes , Dendritic Cells
3.
J Cancer Res Clin Oncol ; 149(16): 14619-14630, 2023 Nov.
Article in English | MEDLINE | ID: mdl-37584709

ABSTRACT

PURPOSE: In a previous study, protein tyrosine phosphatase non-receptor type (PTPN) 3 was identified as an immune checkpoint molecule in lymphocytes, and its potential as a novel target for cancer immunotherapy was anticipated. However, evaluation of dendritic cell (DC) function as antigen-presenting cells is critical for the development of immunotherapy. In this study, we aimed to analyze the biological effect of PTPN3 on DCs induced from human peripheral blood monocytes obtained from healthy individuals. METHODS: We used short-interfering RNA to knock down PTP3 in DCs. For DC maturation, we added cancer cell lysate and tumor necrosis factor-α/interferon-α to immature DCs. In the cytotoxic assay, the target cancer cells were SBC5, unmatched with DCs from healthy human leukocyte antigen (HLA)-A24, or Sq-1, matched with DCs. Enzyme-linked immunosorbent assay was used to determine the amount of cytokines. To examine the intracellular signaling system, intracellular staining was used. RESULTS: PTPN3 knockdown significantly increased the number of DCs, expression of CD80 and chemokine receptor (CCR)7, and production of interleukin-12p40/p70 in mature DCs. In the HLA-A24-restricted DC and human lung squamous cell carcinoma cell cytotoxic assay, inhibition of PTPN3 expression in mature DCs induced cytotoxic T lymphocytes with increased production of INF-γ and granzyme B, and enhanced toxicity against cancer cells and migration to cancer. Furthermore, inhibition of PTPN3 expression activated the mitogen-activated protein kinase pathway in DCs. CONCLUSION: Based on our findings, inhibition of PTPN3 expression could contribute to the development of novel cancer immunotherapies that activate not only lymphocytes but also DCs.


Subject(s)
Dendritic Cells , Neoplasms , Humans , Cytokines/metabolism , T-Lymphocytes, Cytotoxic , Interleukins , Neoplasms/metabolism , Immunotherapy , Protein Tyrosine Phosphatase, Non-Receptor Type 3/metabolism
5.
Immunol Invest ; 51(5): 1498-1514, 2022 Jul.
Article in English | MEDLINE | ID: mdl-34486463

ABSTRACT

Although immune checkpoint inhibitors (ICIs) have emerged as new therapeutic options for refractory cancer, they are only effective in select patients. Tumor antigen-pulsed dendritic cell (DC) vaccine therapy activates tumor-specific cytotoxic T lymphocytes, making it an important immunotherapeutic strategy. Salivary ductal carcinoma (SDC) carries a poor prognosis, including poor long-term survival after metastasis or recurrence. In this study, we reported a case of refractory metastatic SDC that was treated with a tumor lysate-pulsed DC vaccine followed by a single injection of low-dose nivolumab, and a durable complete response was achieved. We retrospectively analyzed the immunological factors that contributed to these long-lasting clinical effects. First, we performed neoantigen analysis using resected metastatic tumor specimens obtained before treatment. We found that the tumor had 256 non-synonymous mutations and 669 class I high-affinity binding neoantigen peptides. Using synthetic neoantigen peptides and ELISpot analysis, we found that peripheral blood mononuclear leukocytes cryopreserved before treatment contained pre-existing neoantigen-specific T cells, and the cells obtained after treatment exhibited greater reactivity to neoantigens than those obtained before treatment. Our results collectively suggest that the rapid and long-lasting effect of this combination therapy in our patient may have resulted from the presence of pre-existing neoantigen-specific T cells and stimulation and expansion of those cells following tumor lysate-pulsed DC vaccine and ICI therapy.


Subject(s)
Cancer Vaccines , Carcinoma, Ductal , Carcinoma , Antigens, Neoplasm , Cancer Vaccines/therapeutic use , Carcinoma, Ductal/therapy , Dendritic Cells , Humans , Leukocytes, Mononuclear , Nivolumab/therapeutic use , Peptides , Retrospective Studies , Salivary Ducts/metabolism
6.
Sci Rep ; 11(1): 13590, 2021 06 30.
Article in English | MEDLINE | ID: mdl-34193879

ABSTRACT

Neoantigens are tumour-specific antigens that arise from non-synonymous mutations in tumour cells. However, their effect on immune responses in the tumour microenvironment remains unclear in breast cancer. We performed whole exome and RNA sequencing of 31 fresh breast cancer tissues and neoantigen prediction from non-synonymous single nucleotide variants (nsSNVs) among exonic mutations. Neoantigen profiles were determined by predictive HLA binding affinity (IC50 < 500 nM) and mRNA expression with a read count of ≥ 1. We evaluated the association between neoantigen load and expression levels of immune-related genes. Moreover, using primary tumour cells established from pleural fluid of a breast cancer patient with carcinomatous pleurisy, we induced cytotoxic T lymphocytes (CTLs) by coculturing neoantigen peptide-pulsed dendritic cells (DCs) with autologous peripheral lymphocytes. The functions of CTLs were examined by cytotoxicity and IFN-γ ELISpot assays. Neoantigen load ranged from 6 to 440 (mean, 95) and was positively correlated to the total number of nsSNVs. Although no associations between neoantigen load and mRNA expression of T cell markers were observed, the coculture of neoantigen-pulsed DCs and lymphocytes successfully induced CTLs ex vivo. These results suggest that neoantigen analysis may have utility in developing strategies to elicit T cell responses.


Subject(s)
Antigens, Neoplasm/immunology , Breast Neoplasms/immunology , Immunity, Cellular , Lymphocytes, Tumor-Infiltrating/immunology , T-Lymphocytes, Cytotoxic/immunology , Adult , Aged , Antigens, Neoplasm/genetics , Breast Neoplasms/genetics , Dendritic Cells/immunology , Female , Humans , Middle Aged , Exome Sequencing
7.
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
8.
Anticancer Res ; 41(8): 4101-4115, 2021 Aug.
Article in English | MEDLINE | ID: mdl-34281881

ABSTRACT

BACKGROUND/AIM: Neoantigens are tumor-specific antigens that emerge due to gene mutations in tumor cells, and are highly antigenic epitopes that escape central immune tolerance in the thymus, making cancer vaccine therapy a desirable option. PATIENTS AND METHODS: Tumor neoantigens were predicted in 17 patients with advanced cancer. They were resistant to the standard treatment regime, and their synthetic peptides were pulsed to the patient's monocyte-derived dendritic cells (DCs), and administered to the patient's lymph nodes via ultrasound. RESULTS: Some patients showed sustained tumor shrinkage after this treatment, while some did not respond, showing no ELISpot reaction. Although the number of mutations and the predicted neoantigen epitopes differed between patients, the clinical effect depended more on the presence or absence of an immune response after vaccination rather than the number of neoantigens. CONCLUSION: Intranodal neoantigen peptide-pulsed DC vaccine administration therapy has clinical and immunological efficacy and safety.


Subject(s)
Antigens, Neoplasm/administration & dosage , Cancer Vaccines/therapeutic use , Dendritic Cells , Neoplasms/therapy , Peptides/administration & dosage , Adult , Aged , Female , Humans , Immunotherapy , Male , Middle Aged , Retrospective Studies , Treatment Outcome
9.
Transl Oncol ; 14(9): 101152, 2021 Sep.
Article in English | MEDLINE | ID: mdl-34134073

ABSTRACT

In our previous study, we found that inhibition of protein tyrosine phosphatase non-receptor type 3 (PTPN3), which is expressed in lymphocytes, enhances lymphocyte activation, suggesting PTPN3 may act as an immune checkpoint molecule. However, PTPN3 is also expressed in various cancers, and the biological significance of PTPN3 in cancer cells is still not well understood, especially for lung neuroendocrine tumor (NET).Therefore, we analyzed the biological significance of PTPN3 in small cell lung cancer and examined the potential for PTPN3 inhibitory treatment as a cancer treatment approach in lung NET including small cell lung cancer (SCLC) and large cell neuroendocrine cancer (LCNEC). Experiments in a mouse xenograft model using allo lymphocytes showed that PTPN3 inhibition in SCLC cells enhanced the anti-tumor effect of PTPN3-suppressed activated lymphocytes. In addition, PTPN3 was associated with increased vascularization, decreased CD8/FOXP3 ratio and cellular immunosuppression in SCLC clinical specimens. Experiments in a mouse xenograft model using autocrine lymphocytes also showed that PTPN3 inhibition in LCNEC cells augmented the anti-tumor effect of PTPN3-suppressed activated lymphocytes. In vitro experiments showed that PTPN3 is involved in the induction of malignant traits such as proliferation, invasion and migration. Signaling from PTPN3 is mediated by MAPK and PI3K signals via tyrosine kinase phosphorylation through CACNA1G calcium channel. Our results show that PTPN3 suppression is associated with lymphocyte activation and cancer suppression in lung NET. These results suggest that PTPN3 suppression could be a new method of cancer treatment and a major step in the development of new cancer immunotherapies.

10.
Anticancer Res ; 40(8): 4663-4674, 2020 Aug.
Article in English | MEDLINE | ID: mdl-32727790

ABSTRACT

BACKGROUND/AIM: Roles for mutant (mt) KRAS in the innate immune microenvironment in colorectal cancer (CRC) were explored. MATERIALS AND METHODS: Human CRC HCT116-derived, mtKRAS-disrupted (HKe3) cells that express exogenous mtKRAS and allogenic cytokine-activated killer (CAK) cells were co-cultured in 3D floating (3DF) culture. The anti-CD155 antibody was used for function blocking and immuno histochemistry. RESULTS: Infiltration of CAK cells, including NKG2D+ T cells, into the deep layer of HKe3-mtKRAS spheroids, was observed. Surface expression of CD155 was found to be up-regulated by mtKRAS in 3DF culture and CRC tissues. Further, the number of CD3+ tumor-infiltrating cells in the invasion front that show substantial CD155 expression was significantly larger than the number showing weak expression in CRC tissues with mtKRAS. CD155 blockade decreased the growth of spheroids directly and indirectly through the release of CAK cells. CONCLUSION: CD155 blockade may be useful for therapies targeting tumors containing mtKRAS.


Subject(s)
Immune Evasion/immunology , NK Cell Lectin-Like Receptor Subfamily K/immunology , Proto-Oncogene Proteins p21(ras)/immunology , Receptors, Virus/immunology , T-Lymphocytes/immunology , Aged , Aged, 80 and over , Cell Line , Cell Line, Tumor , Coculture Techniques/methods , Colorectal Neoplasms/immunology , Female , Humans , Killer Cells, Natural/immunology , Male , Middle Aged , Tumor Microenvironment/immunology
11.
J Immunother ; 43(4): 121-133, 2020 05.
Article in English | MEDLINE | ID: mdl-31834207

ABSTRACT

Pancreatic ductal adenocarcinoma (PDAC) is resistant to immunotherapy. As a factor of resistance, the dense fibrosis of this cancer acts as a barrier to inhibit immune cell infiltration into a tumor. We examined the influence of a Hedgehog signal inhibitor, Patched 1-interacting peptide, on fibrosis, infiltration of immune cells, and immunotherapeutic effects on PDAC. We found that this peptide inhibited proliferation and migration of cancer-associated fibroblasts and cancer cells. Furthermore, this peptide reduced the production of extracellular matrix and transforming growth factor ß1 in cancer-associated fibroblasts and induced expression of HLA-ABC in PDAC cells and interferon-γ in lymphocytes. In vivo, the peptide suppressed fibrosis of PDAC and increased immune cell infiltration into tumors. The combination of this peptide and an anti-programmed death-1 antibody augmented the antitumor effect, and this combination showed the same effect in experiments using cancer cells and autologous lymphocytes. These results indicate that, in addition to the direct effect of tumor suppression, the Patched 1-interacting peptide increases the infiltration of immune cells by reducing fibrosis of PDAC and consequently enhances the effect of immunotherapy. Therefore, treatment with this peptide may be a novel therapy with 2 different mechanisms: direct tumor suppression and enhancing the immune response against PDAC.


Subject(s)
Pancreatic Neoplasms/etiology , Pancreatic Neoplasms/pathology , Patched-1 Receptor/metabolism , Peptides/metabolism , Protein Interaction Domains and Motifs , Animals , Antineoplastic Agents, Immunological/pharmacology , Carcinoma, Pancreatic Ductal/etiology , Carcinoma, Pancreatic Ductal/metabolism , Carcinoma, Pancreatic Ductal/pathology , Carcinoma, Pancreatic Ductal/therapy , Cell Communication , Cell Line, Tumor , Disease Models, Animal , Disease Susceptibility , Fibrosis , Humans , Immunotherapy , Lymphocyte Activation/drug effects , Lymphocyte Activation/genetics , Lymphocyte Activation/immunology , Lymphocytes/drug effects , Lymphocytes/immunology , Lymphocytes/metabolism , Lymphocytes, Tumor-Infiltrating/immunology , Lymphocytes, Tumor-Infiltrating/metabolism , Lymphocytes, Tumor-Infiltrating/pathology , Mice , Pancreatic Neoplasms/metabolism , Pancreatic Neoplasms/therapy , Patched-1 Receptor/chemistry , Patched-1 Receptor/genetics , Peptides/pharmacology , Tumor Microenvironment/drug effects , Tumor Microenvironment/genetics , Tumor Microenvironment/immunology , Xenograft Model Antitumor Assays
12.
Cancer Immunol Immunother ; 68(10): 1649-1660, 2019 Oct.
Article in English | MEDLINE | ID: mdl-31562536

ABSTRACT

It has been shown that protein tyrosine phosphatase non-receptor type (PTPN) 3 inhibits T-cell activation. However, there is no definitive conclusion about how the inhibition of PTPN3 in lymphocytes affects immune functions in human lymphocytes. In the present study, we showed that PTPN3 inhibition significantly contributes to the enhanced activation of activated human lymphocytes. The PTPN3 expression of lymphocytes was significantly increased through the activation process using IL-2 and anti-CD3 mAb. Interestingly, inhibiting the PTPN3 expression in activated lymphocytes significantly augmented the proliferation, migration, and cytotoxicity through the phosphorylation of zeta-chain-associated protein kinase 70 (ZAP-70), lymphocyte-specific protein tyrosine kinase (LCK), and extracellular signal-regulated kinases (ERK). Lymphocyte activation by PTPN3 inhibition was observed only in activated CD3+ T cells and not in NK cells or resting T cells. In therapy experiments using autologous tumors and lymphocytes, PTPN3 inhibition significantly augmented the number of tumor-infiltrated lymphocytes and the cytotoxicity of activated lymphocytes. Our results strongly imply that PTPN3 acts as an immune checkpoint in activated lymphocytes and that PTPN3 inhibitor may be a new non-antibody-type immune checkpoint inhibitor for cancer therapy.


Subject(s)
Cell Cycle Checkpoints , Lymphocyte Activation , Ovarian Neoplasms/prevention & control , Protein Tyrosine Phosphatase, Non-Receptor Type 3/antagonists & inhibitors , T-Lymphocytes/immunology , Animals , Apoptosis , Cell Movement , Cell Proliferation , Female , Humans , Lymphocyte Specific Protein Tyrosine Kinase p56(lck)/metabolism , Mice , Mice, Inbred NOD , Mice, SCID , Ovarian Neoplasms/immunology , Ovarian Neoplasms/metabolism , Ovarian Neoplasms/pathology , Phosphorylation , Receptors, Antigen, T-Cell/metabolism , Signal Transduction , Tumor Cells, Cultured , Xenograft Model Antitumor Assays , ZAP-70 Protein-Tyrosine Kinase/metabolism
13.
Anticancer Res ; 39(8): 4517-4523, 2019 Aug.
Article in English | MEDLINE | ID: mdl-31366554

ABSTRACT

BACKGROUND/AIM: Immune checkpoint inhibitors (ICIs) have dramatically changed the clinical outcomes of advanced tumours. However, biomarkers for monitoring immunological features during immunotherapy remain unclear, especially those in the peripheral blood, which are easily available. This study evaluated the usefulness of nCounter Analysis System in identifying immunological biomarkers in peripheral blood mononuclear cells (PBMCs) during ICI therapy. PATIENTS AND METHODS: PBMCs from two patients who responded well to ICI therapy were used, and the expression levels of immune-related mRNA and extracellular proteins were analyzed. RESULTS: Changes in the expression levels of 55 genes from pre-treatment to on-treatment were bioinformatically similar between the two cases. The expression levels of PD-1 were consistent with those by flow cytometry analysis, a reliable tool for monitoring various markers. CONCLUSION: The nCounter Analysis System may be a potent tool to simultaneously investigate genes and proteins on PBMCs as biomarkers during immunotherapy using a small amount of sample.


Subject(s)
Biomarkers, Tumor/blood , Immunotherapy , Lung Neoplasms/blood , Neoplasm Proteins/genetics , Aged , Gene Expression Regulation, Neoplastic/immunology , Humans , Leukocytes, Mononuclear/immunology , Leukocytes, Mononuclear/metabolism , Lung Neoplasms/genetics , Lung Neoplasms/immunology , Lung Neoplasms/therapy , Male , Middle Aged , RNA, Messenger/blood
14.
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
15.
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
16.
Anticancer Res ; 38(7): 4273-4279, 2018 Jul.
Article in English | MEDLINE | ID: mdl-29970561

ABSTRACT

BACKGROUND/AIM: Epithelial cell adhesion molecule (EpCAM) is expressed in various types of cancer, including breast cancer, and is correlated with metastasis, invasion, therapeutic resistance and prognosis. Moreover, several cell surface markers, such as CD44 and EpCAM, are molecular targets on cancer stem-like cells of breast cancer. The aim of this study was to investigate whether catumaxomab, a clinical-grade bispecific antibody that binds to both EpCAM on tumor cells and CD3 on T-cells, combined with activated T-cells can eliminate chemoresistant triple-negative breast cancer (TNBC) cells in vitro. MATERIALS AND METHODS: First, a cell line (MUK-BC1) was established from human breast carcinoma cells derived from a patient with chemoresistant and disseminated breast cancer. These EpCAM-positive TNBC cells were almost completely resistant to various drug-mediated cytotoxicities up to a concentration of 10 µg/ml. RESULTS: Pre-treatment with catumaxomab and subsequent addition of interleukin-2/OKT3-activated autologous T-cells eliminated EpCAM-positive TNBC cells. CONCLUSION: Catumaxomab combined with activated T-cells may be a potent therapeutic modality to overcome chemoresistant EpCAM-positive TNBC cells.


Subject(s)
Antibodies, Bispecific/pharmacology , Antineoplastic Agents, Immunological/pharmacology , Immunotherapy/methods , T-Lymphocytes/immunology , Triple Negative Breast Neoplasms , Adult , Cell Line, Tumor , Epithelial Cell Adhesion Molecule/antagonists & inhibitors , Female , Humans , Lymphocyte Activation
17.
Anticancer Res ; 38(5): 2739-2748, 2018 05.
Article in English | MEDLINE | ID: mdl-29715094

ABSTRACT

BACKGROUND/AIM: Metronidazole (MNZ) is a common antibiotic that exerts disulfiram-like effects when taken together with alcohol. However, the relationship between MNZ and aldehyde dehydrogenase (ALDH) activity remains unclear. This study investigated whether MNZ reduces cancer stemness by suppressing ALDH activity and accordingly reducing the malignancy of cholangiocarcinoma (CCA). MATERIALS AND METHODS: We developed gemcitabine (GEM)-resistant TFK-1 cells and originally established CCA cell line from a patient with GEM-resistant CCA. Using these cell lines, we analyzed the impacts of MNZ for cancer stem cell markers, invasiveness, and chemosensitivity. RESULTS: MNZ reduced ALDH activity in GEM-resistant CCA cells, leading to decreased invasiveness and enhanced chemosensitivity. MNZ diminished the invasiveness by inducing mesenchymal-epithelial transition and enhancing chemosensitivity by increasing ENT1 (equilibrative nucleoside transporter 1) and reducing RRM1 (ribonucleotide reductase M1). CONCLUSION: MNZ reduced cancer stemness in GEM-resistant CCA cells. Combined GEM and MNZ would be a promising therapeutic strategy for cancer stem-like CAA.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Bile Duct Neoplasms/drug therapy , Cholangiocarcinoma/drug therapy , Neoplastic Stem Cells/drug effects , Aldehyde Dehydrogenase/biosynthesis , Aldehyde Dehydrogenase/genetics , Bile Duct Neoplasms/metabolism , Biomarkers, Tumor , Cell Line, Tumor , Cell Transdifferentiation/drug effects , Cholangiocarcinoma/metabolism , Deoxycytidine/administration & dosage , Deoxycytidine/analogs & derivatives , Drug Resistance, Neoplasm , Drug Screening Assays, Antitumor , Humans , Male , Metronidazole/administration & dosage , Middle Aged , Neoplasm Invasiveness , Neoplasm Proteins/biosynthesis , Neoplasm Proteins/genetics , Neoplastic Stem Cells/enzymology , Spheroids, Cellular/drug effects , Gemcitabine
18.
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
19.
Anticancer Res ; 36(7): 3585-9, 2016 Jul.
Article in English | MEDLINE | ID: mdl-27354627

ABSTRACT

BACKGROUND/AIM: The serine/threonine-protein kinase B-Raf (BRAF) V600E mutant (BRAF(V600E)) inhibitor vemurafenib, has improved clinical outcomes for patients with BRAF(V600E) melanoma, but acquired cellular resistance mediated by AKT serine/threonine kinase 1 (AKT) phosphorylation limits its efficacy. We examined the effect of resveratrol on vemurafenib-resistant melanoma cells. MATERIALS AND METHODS: A vemurafenib-resistant human metastatic melanoma cell line positive for the BRAF V600E mutation was established. The anti-tumorigenic effects of vemurafenib and resveratrol, both alone and in combination, were examined through analysis of cell proliferation and protein expression. RESULTS: The level of phosphorylated AKT (p-AKT) was increased in the primary melanoma cells after treatment with vemurafenib, and the basal level of p-AKT was increased in vemurafenib-resistant melanoma cells. Notably, resveratrol both alone and in combination with vemurafenib effectively suppressed cell proliferation and AKT phosphorylation in both parental and vemurafenib-resistant melanoma cells. CONCLUSION: Vemurafenib resistance can be reversed by addition of resveratrol in patients undergoing treatment with BRAF inhibitors.


Subject(s)
Antineoplastic Agents/pharmacology , Indoles/pharmacology , Protein Processing, Post-Translational/drug effects , Proto-Oncogene Proteins B-raf/genetics , Proto-Oncogene Proteins c-akt/metabolism , Stilbenes/pharmacology , Sulfonamides/pharmacology , Drug Resistance, Neoplasm , Drug Synergism , Humans , Inhibitory Concentration 50 , Melanoma/drug therapy , Melanoma/pathology , Mutation, Missense , Phosphorylation , Resveratrol , Tumor Cells, Cultured , Vemurafenib
20.
Anticancer Res ; 36(2): 653-7, 2016 Feb.
Article in English | MEDLINE | ID: mdl-26851020

ABSTRACT

Development of an effective therapeutic strategy for refractory pancreatic cancer must consider whether chemosensitivity can be induced in chemoresistant cells. We established a pancreatic cancer stem cell-rich cell line using TIG-1 feeder cells and leukemia inhibitory factor (LIF)-rich SNL76/7 conditioned medium. We generated a cell line, namely YNPC031312-B, following isolation of cells from the malignant ascites of a patient with gemcitabine-resistant pancreatic cancer. A YNPC031312-B-Hypoxia cell line was established by maintaining YNPC031312-B cells under tumor-like hypoxic conditions (1% O2). Both cell lines exhibited a pancreatic cancer stem cell phenotype: YNPC031312-B cells were CD24(+)CD44(-)CD133(+)epithelial cell adhesion molecule (EpCAM)(+)alkaline phosphatase(+)Octamer-binding transcription factor (OCT)3/4+and YNPC031312-B-Hypoxia cells were CD24(+)CD44(+)CD133(+)EpCAM(+). YNPC031312-B-Hypoxia cells were larger, had superior migratory ability, and higher gemcitabine sensitivity compared to YNPC031312-B cells. The use of LIF or other factors with similar bioactivity under hypoxic conditions may contribute to the phenotypic change to gemcitabine sensitivity. Our results may aid development of new therapeutic strategies targeting refractory pancreatic cancer.


Subject(s)
Deoxycytidine/analogs & derivatives , Drug Resistance, Neoplasm , Hypoxia , Leukemia Inhibitory Factor/metabolism , Mediator Complex/metabolism , Neoplastic Stem Cells/drug effects , Pancreatic Neoplasms/drug therapy , Antimetabolites, Antineoplastic/pharmacology , Apoptosis/drug effects , Blotting, Western , Cell Movement/drug effects , Cell Proliferation/drug effects , Deoxycytidine/pharmacology , Epithelial-Mesenchymal Transition/drug effects , Humans , Immunoenzyme Techniques , Neoplastic Stem Cells/metabolism , Neoplastic Stem Cells/pathology , Pancreatic Neoplasms/metabolism , Pancreatic Neoplasms/pathology , Tumor Cells, Cultured , Gemcitabine
SELECTION OF CITATIONS
SEARCH DETAIL
...