Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 178
Filter
Add more filters

Country/Region as subject
Publication year range
1.
Br J Cancer ; 130(10): 1647-1658, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38555315

ABSTRACT

BACKGROUND: Cancer-associated fibroblasts (CAFs) in the tumor microenvironment (TME) play a critical role in tumor immunosuppression. However, targeted depletion of CAFs is difficult due to their diverse cells of origin and the resulting lack of specific surface markers. Near-infrared photoimmunotherapy (NIR-PIT) is a novel cancer treatment that leads to rapid cell membrane damage. METHODS: In this study, we used anti-mouse fibroblast activation protein (FAP) antibody to target FAP+ CAFs (FAP-targeted NIR-PIT) and investigated whether this therapy could suppress tumor progression and improve tumor immunity. RESULTS: FAP-targeted NIR-PIT induced specific cell death in CAFs without damaging adjacent normal cells. Furthermore, FAP-targeted NIR-PIT treated mice showed significant tumor regression in the CAF-rich tumor model accompanied by an increase in CD8+ tumor infiltrating lymphocytes (TILs). Moreover, treated tumors showed increased levels of IFN-γ, TNF-α, and IL-2 in CD8+ TILs compared with non-treated tumors, suggesting enhanced antitumor immunity. CONCLUSIONS: Cancers with FAP-positive CAFs in their TME grow rapidly and FAP-targeted NIR-PIT not only suppresses their growth but improves tumor immunosuppression. Thus, FAP-targeted NIR-PIT is a potential therapeutic strategy for selectively targeting the TME of CAF+ tumors.


Subject(s)
Cancer-Associated Fibroblasts , Immunotherapy , Tumor Microenvironment , Animals , Cancer-Associated Fibroblasts/immunology , Cancer-Associated Fibroblasts/metabolism , Mice , Immunotherapy/methods , Tumor Microenvironment/immunology , Endopeptidases , Serine Endopeptidases/metabolism , Gelatinases/metabolism , Membrane Proteins/metabolism , Lymphocytes, Tumor-Infiltrating/immunology , Female , Humans , Infrared Rays/therapeutic use , Phototherapy/methods , CD8-Positive T-Lymphocytes/immunology , Cell Line, Tumor , Mice, Inbred C57BL
2.
Br J Cancer ; 130(7): 1187-1195, 2024 Apr.
Article in English | MEDLINE | ID: mdl-38316993

ABSTRACT

BACKGROUND: Pancreatic cancer is an aggressive, immunologically "cold" tumor. Oncolytic virotherapy is a promising treatment to overcome this problem. We developed a telomerase-specific oncolytic adenovirus armed with p53 gene (OBP-702). METHODS: We investigated the efficacy of OBP-702 for pancreatic cancer, focusing on its long-term effects via long-lived memory CD8 + T cells including tissue-resident memory T cells (TRMs) and effector memory T cells (TEMs) differentiated from effector memory precursor cells (TEMps). RESULTS: First, in vitro, OBP-702 significantly induced adenosine triphosphate (ATP), which is important for memory T cell establishment. Next, in vivo, OBP-702 local treatment to murine pancreatic PAN02 tumors increased TEMps via ATP induction from tumors and IL-15Rα induction from macrophages, leading to TRM and TEM induction. Activation of these memory T cells by OBP-702 was also maintained in combination with gemcitabine+nab-paclitaxel (GN) in a PAN02 bilateral tumor model, and GN + OBP-702 showed significant anti-tumor effects and increased TRMs in OBP-702-uninjected tumors. Finally, in a neoadjuvant model, in which PAN02 cells were re-inoculated after resection of treated-PAN02 tumors, GN + OBP-702 provided long-term anti-tumor effects even after tumor resection. CONCLUSION: OBP-702 can be a long-term immunostimulant with sustained anti-tumor effects on immunologically cold pancreatic cancer.


Subject(s)
Oncolytic Virotherapy , Oncolytic Viruses , Pancreatic Neoplasms , Telomerase , Humans , Animals , Mice , Adenoviridae/genetics , Adenoviridae/metabolism , Tumor Suppressor Protein p53/genetics , Telomerase/genetics , Telomerase/metabolism , Cell Line, Tumor , Pancreatic Neoplasms/genetics , Pancreatic Neoplasms/therapy , Adenosine Triphosphate , Oncolytic Viruses/genetics , Oncolytic Viruses/metabolism
3.
Acta Med Okayama ; 78(2): 151-161, 2024 Apr.
Article in English | MEDLINE | ID: mdl-38688833

ABSTRACT

Soft-tissue sarcoma (STS) is a heterogeneous group of rare tumors originating predominantly from the embryonic mesoderm. Despite the development of combined modalities including radiotherapy, STSs are often refractory to antitumor modalities, and novel strategies that improve the prognosis of STS patients are needed. We previously demonstrated the therapeutic potential of two telomerase-specific replication-competent oncolytic adenoviruses, OBP-301 and tumor suppressor p53-armed OBP-702, in human STS cells. Here, we demonstrate in vitro and in vivo antitumor effects of OBP-702 in combination with ionizing radiation against human STS cells (HT1080, NMS-2, SYO-1). OBP-702 synergistically promoted the antitumor effect of ionizing radiation in the STS cells by suppressing the expression of B-cell lymphoma-X large (BCL-xL) and enhancing ionizing radiation-induced apoptosis. The in vivo experiments demonstrated that this combination therapy significantly suppressed STS tumors' growth. Our results suggest that OBP-702 is a promising antitumor reagent for promoting the radiosensitivity of STS tumors.


Subject(s)
Oncolytic Virotherapy , Radiation Tolerance , Sarcoma , Tumor Suppressor Protein p53 , bcl-X Protein , Sarcoma/therapy , Sarcoma/radiotherapy , Humans , Oncolytic Virotherapy/methods , bcl-X Protein/genetics , bcl-X Protein/metabolism , Cell Line, Tumor , Animals , Tumor Suppressor Protein p53/genetics , Tumor Suppressor Protein p53/metabolism , Mice , Apoptosis , Adenoviridae/genetics
4.
Cancer Immunol Immunother ; 72(5): 1285-1300, 2023 May.
Article in English | MEDLINE | ID: mdl-36436021

ABSTRACT

Pancreatic ductal adenocarcinoma (PDAC) is often refractory to treatment with gemcitabine (GEM) and immune checkpoint inhibitors including anti-programmed cell death ligand 1 (PD-L1) antibody. However, the precise relationship between GEM-resistant PDAC and development of an immunosuppressive tumor microenvironment (TME) remains unclear. In this study, we investigated the immunosuppressive TME in parental and GEM-resistant PDAC tumors and assessed the therapeutic potential of combination therapy with the telomerase-specific replication-competent oncolytic adenovirus OBP-702, which induces tumor suppressor p53 protein and PD-L1 blockade against GEM-resistant PDAC tumors. Mouse PDAC cells (PAN02) and human PDAC cells (MIA PaCa-2, BxPC-3) were used to establish GEM-resistant PDAC lines. PD-L1 expression and the immunosuppressive TME were analyzed using parental and GEM-resistant PDAC cells. A cytokine array was used to investigate the underlying mechanism of immunosuppressive TME induction by GEM-resistant PAN02 cells. The GEM-resistant PAN02 tumor model was used to evaluate the antitumor effect of combination therapy with OBP-702 and PD-L1 blockade. GEM-resistant PDAC cells exhibited higher PD-L1 expression and produced higher granulocyte-macrophage colony-stimulating factor (GM-CSF) levels compared with parental cells, inducing an immunosuppressive TME and the accumulation of myeloid-derived suppressor cells (MDSCs). OBP-702 significantly inhibited GEM-resistant PAN02 tumor growth by suppressing GM-CSF-mediated MDSC accumulation. Moreover, combination treatment with OBP-702 significantly enhanced the antitumor efficacy of PD-L1 blockade against GEM-resistant PAN02 tumors. The present results suggest that combination therapy involving OBP-702 and PD-L1 blockade is a promising antitumor strategy for treating GEM-resistant PDAC with GM-CSF-induced immunosuppressive TME formation.


Subject(s)
Carcinoma, Pancreatic Ductal , Myeloid-Derived Suppressor Cells , Oncolytic Viruses , Pancreatic Neoplasms , Mice , Animals , Humans , Gemcitabine , Myeloid-Derived Suppressor Cells/metabolism , Granulocyte-Macrophage Colony-Stimulating Factor/pharmacology , B7-H1 Antigen/metabolism , Carcinoma, Pancreatic Ductal/metabolism , Immunosuppressive Agents/therapeutic use , Tumor Microenvironment , Cell Line, Tumor , Pancreatic Neoplasms
5.
Cancer Immunol Immunother ; 72(7): 2029-2044, 2023 Jul.
Article in English | MEDLINE | ID: mdl-36764954

ABSTRACT

Cancer-associated fibroblasts (CAFs) are a critical component of the tumor microenvironment and play a central role in tumor progression. Previously, we reported that CAFs might induce tumor immunosuppression via interleukin-6 (IL-6) and promote tumor progression by blocking local IL-6 in the tumor microenvironment with neutralizing antibody. Here, we explore whether an anti-IL-6 receptor antibody could be used as systemic therapy to treat cancer, and further investigate the mechanisms by which IL-6 induces tumor immunosuppression. In clinical samples, IL-6 expression was significantly correlated with α-smooth muscle actin expression, and high IL-6 cases showed tumor immunosuppression. Multivariate analysis showed that IL-6 expression was an independent prognostic factor. In vitro, IL-6 contributed to cell proliferation and differentiation into CAFs. Moreover, IL-6 increased hypoxia-inducible factor 1α (HIF1α) expression and induced tumor immunosuppression by enhancing glucose uptake by cancer cells and competing for glucose with immune cells. MR16-1, a rodent analog of anti-IL-6 receptor antibody, overcame CAF-induced immunosuppression and suppressed tumor progression in immunocompetent murine cancer models by regulating HIF1α activation in vivo. The anti-IL-6 receptor antibody could be systemically employed to overcome tumor immunosuppression and improve patient survival with various cancers. Furthermore, the tumor immunosuppression was suggested to be induced by IL-6 via HIF1α activation.


Subject(s)
Cancer-Associated Fibroblasts , Carcinoma, Squamous Cell , Animals , Mice , Cancer-Associated Fibroblasts/pathology , Carcinoma, Squamous Cell/pathology , Interleukin-6/metabolism , Immune Tolerance , Immunosuppression Therapy , Tumor Microenvironment , Cell Line, Tumor
6.
Cancer Immunol Immunother ; 72(11): 3787-3802, 2023 Nov.
Article in English | MEDLINE | ID: mdl-37668710

ABSTRACT

The programmed cell death 1 protein (PD-1)/programmed cell death ligand 1 (PD-L1) axis plays a crucial role in tumor immunosuppression, while the cancer-associated fibroblasts (CAFs) have various tumor-promoting functions. To determine the advantage of immunotherapy, the relationship between the cancer cells and the CAFs was evaluated in terms of the PD-1/PD-L1 axis. Overall, 140 cases of esophageal cancer underwent an immunohistochemical analysis of the PD-L1 expression and its association with the expression of the α smooth muscle actin, fibroblast activation protein, CD8, and forkhead box P3 (FoxP3) positive cells. The relationship between the cancer cells and the CAFs was evaluated in vitro, and the effect of the anti-PD-L1 antibody was evaluated using a syngeneic mouse model. A survival analysis showed that the PD-L1+ CAF group had worse survival than the PD-L1- group. In vitro and in vivo, direct interaction between the cancer cells and the CAFs showed a mutually upregulated PD-L1 expression. In vivo, the anti-PD-L1 antibody increased the number of dead CAFs and cancer cells, resulting in increased CD8+ T cells and decreased FoxP3+ regulatory T cells. We demonstrated that the PD-L1-expressing CAFs lead to poor outcomes in patients with esophageal cancer. The cancer cells and the CAFs mutually enhanced the PD-L1 expression and induced tumor immunosuppression. Therefore, the PD-L1-expressing CAFs may be good targets for cancer therapy, inhibiting tumor progression and improving host tumor immunity.


Subject(s)
Cancer-Associated Fibroblasts , Esophageal Neoplasms , Animals , Mice , Humans , B7-H1 Antigen/metabolism , Cancer-Associated Fibroblasts/pathology , CD8-Positive T-Lymphocytes , Programmed Cell Death 1 Receptor/metabolism , Immunosuppression Therapy , Forkhead Transcription Factors/metabolism , Tumor Microenvironment
7.
Acta Med Okayama ; 77(3): 311-318, 2023 Jun.
Article in English | MEDLINE | ID: mdl-37357632

ABSTRACT

Japan's hip fracture management guidelines now recommend the use of cemented stems in cases of bone fragility. However, the current stem selection practices in bipolar hemiarthroplasty (BHA) in a super-aging area in Japan remain unclear. This study aimed to examine the stem selection policies, the surgeons' concerns about cemented stems, and factors affecting their confidence in their ability to coach others on cemented stem procedures. Ninety-four orthopedic surgeons (27 facilities) responded to our web-based questionnaire conducted in January/February 2022. Cementless stem was the first choice of 97.8% of the surgeons; <15% of the respondents expected to increase their use of cemented stems in the future. The cement technique was the greatest concern; almost half of the surgeons described having insufficient experience with cemented stems. The factor that most affected the surgeons' expertise in using cemented stems is the number of surgeries they had conducted with a cemented stem (multivariable analysis odds ratio 8.42, p=0.001). Greater experience was associated with increased expertise of the surgeons in using cemented stems, with a threshold of 11 cases showing sensitivity of 41.7% and specificity of 98.3% for their confidence to instruct cemented stems.


Subject(s)
Arthroplasty, Replacement, Hip , Hemiarthroplasty , Hip Fractures , Hip Prosthesis , Surgeons , Humans , Aged , Arthroplasty, Replacement, Hip/methods , Hemiarthroplasty/methods , Japan , Reoperation , Prosthesis Design , Hip Fractures/surgery , Bone Cements , Surveys and Questionnaires
8.
Gan To Kagaku Ryoho ; 50(10): 1102-1103, 2023 Oct.
Article in Japanese | MEDLINE | ID: mdl-38035844

ABSTRACT

Pancreatic ductal adenocarcinoma(PDAC)is lethal malignancy with abundant stroma. Cancer-associated fibroblasts (CAFs) exist in the PDAC stroma and contribute to progression of malignant transformation, treatment resistance, and recurrence. However, effective treatment to control PDAC stroma has not been established. We have developed tumor suppressor gene p53-armed oncolytic adenovirus(OBP-702), and have clarified therapeutic effects on PDAC cells. In this study, we investigate the therapeutic effect of OBP-702 on PDAC CAF.


Subject(s)
Carcinoma, Pancreatic Ductal , Pancreatic Neoplasms , Humans , Adenoviridae/genetics , Pancreatic Neoplasms/drug therapy , Carcinoma, Pancreatic Ductal/drug therapy , Pancreas/pathology , Tumor Microenvironment , Pancreatic Neoplasms
9.
Cancer Sci ; 113(6): 1919-1929, 2022 Jun.
Article in English | MEDLINE | ID: mdl-35398956

ABSTRACT

Conventional imaging techniques are available for clinical identification of tumor sites. However, detecting metastatic tumor cells that are spreading from primary tumor sites using conventional imaging techniques remains difficult. In contrast, fluorescence-based labeling systems are useful tools for detecting tumor cells at the single-cell level in cancer research. The ability to detect fluorescent-labeled tumor cells enables investigations of the biodistribution of tumor cells for the diagnosis and treatment of cancer. For example, the presence of fluorescent tumor cells in the peripheral blood of cancer patients is a predictive biomarker for early diagnosis of distant metastasis. The elimination of fluorescent tumor cells without damaging normal tissues is ideal for minimally invasive treatment of cancer. To capture fluorescent tumor cells within normal tissues, however, tumor-specific activated target molecules are needed. This review focuses on recent advances in tumor-targeted fluorescence labeling systems, in which indirect reporter labeling using tumor-specific promoters is applied to fluorescence labeling of tumor cells for the diagnosis and treatment of cancer. Telomerase promoter-dependent fluorescence labeling using replication-competent viral vectors produces fluorescent proteins that can be used to detect and eliminate telomerase-positive tumor cells. Tissue-specific promoter-dependent fluorescence labeling enables identification of specific tumor cells. Vimentin promoter-dependent fluorescence labeling is a useful tool for identifying tumor cells that undergo epithelial-mesenchymal transition (EMT). The evaluation of tumor cells undergoing EMT is important for accurately assessing metastatic potential. Thus, tumor-targeted fluorescence labeling systems represent novel platforms that enable the capture of tumor cells for the diagnosis and treatment of cancer.


Subject(s)
Neoplasms , Telomerase , Cell Line, Tumor , Epithelial-Mesenchymal Transition , Fluorescence , Humans , Neoplasms/diagnosis , Neoplasms/therapy , Telomerase/metabolism , Tissue Distribution
10.
Br J Cancer ; 127(4): 757-765, 2022 09.
Article in English | MEDLINE | ID: mdl-35597869

ABSTRACT

BACKGROUND: Emerging evidence indicates that immunogenicity plays an important role in intrahepatic cholangiocarcinoma (ICC). Herein, we systematically evaluated the clinical relevance of immunogenicity in ICC. METHODS: Highly immunogenic ICCs identified in the public dataset and the Cancer Immunome Atlas (TCIA) were assessed to determine the prognostic impact of immunogenicity in ICC and key components after curative resection. We also investigated the clinical relevance of the immune milieu in ICC. RESULTS: Using the Gene Expression Omnibus dataset 89749 and TCIA, we identified CD8+/forkhead box P3 (FoxP3)+ tumour-infiltrating lymphocytes (TILs), T-cell immunoglobulin and mucin domain 3 (TIM-3) and human leukocyte antigen-A (HLA-A) in highly immunogenic ICCs. Immunohistochemical analysis of the in-house cohort showed that intratumoral FoxP3+ TILs correlated with CD8+ TILs (P = 0.045, Fisher's exact test) and that high FoxP3+/CD8+ ratio (FCR) was an important marker for poor survival (P < 0.001, log-rank test). Furthermore, the FCR was higher in tumour-free lymph nodes in ICCs with lymph node metastases than in those without lymph node metastases (P = 0.003, Mann-Whitney U test). CONCLUSIONS: FCR should be considered an important biomarker that represents the immune environment of ICC based on its potentially important role in tumour progression, especially lymph node metastasis.


Subject(s)
Bile Duct Neoplasms , Cholangiocarcinoma , Bile Duct Neoplasms/genetics , Bile Duct Neoplasms/pathology , Bile Ducts, Intrahepatic/pathology , Cholangiocarcinoma/genetics , Cholangiocarcinoma/pathology , Forkhead Transcription Factors/genetics , Humans , Lymphatic Metastasis/pathology , Lymphocytes, Tumor-Infiltrating , Prognosis , T-Lymphocytes, Regulatory
11.
Mol Ther ; 29(10): 2920-2930, 2021 10 06.
Article in English | MEDLINE | ID: mdl-34023506

ABSTRACT

Extracellular vesicles (EVs) play important roles in various intercellular communication processes. The abscopal effect is an interesting phenomenon in cancer treatment, in which immune activation is generally considered a main factor. We previously developed a telomerase-specific oncolytic adenovirus, Telomelysin (OBP-301), and occasionally observed therapeutic effects on distal tumors after local treatment in immunodeficient mice. In this study, we hypothesized that EVs may be involved in the abscopal effect of OBP-301. EVs isolated from the supernatant of HCT116 human colon carcinoma cells treated with OBP-301 were confirmed to contain OBP-301, and they showed cytotoxic activity (apoptosis and autophagy) similar to OBP-301. In bilateral subcutaneous HCT116 and CT26 tumor models, intratumoral administration of OBP-301 produced potent antitumor effects on tumors that were not directly treated with OBP-301, involving direct mediation by tumor-derived EVs containing OBP-301. This indicates that immune activation is not the main factor in this abscopal effect. Moreover, tumor-derived EVs exhibited high tumor tropism in orthotopic HCT116 rectal tumors, in which adenovirus E1A and adenovirus type 5 proteins were observed in metastatic liver tumors after localized rectal tumor treatment. In conclusion, local treatment with OBP-301 has the potential to produce abscopal effects via tumor-derived EVs.


Subject(s)
Adenoviridae/genetics , Colonic Neoplasms/therapy , Extracellular Vesicles/transplantation , Liver Neoplasms/secondary , Liver Neoplasms/therapy , Animals , Cell Line, Tumor , Cell Survival , Extracellular Vesicles/virology , HCT116 Cells , Humans , Mice , Oncolytic Viruses/genetics , Viral Tropism , Xenograft Model Antitumor Assays
12.
Acta Med Okayama ; 76(2): 203-215, 2022 Apr.
Article in English | MEDLINE | ID: mdl-35503449

ABSTRACT

The epithelial-mesenchymal transition (EMT), a normal biological process by which epithelial cells acquire a mesenchymal phenotype, is associated with migration, metastasis, and chemoresistance in cancer cells, and with poor prognosis in patients with esophageal cancer. However, therapeutic strategies to inhibit EMT in tumor environments remain elusive. Here, we show the therapeutic potential of telomerase-specific replication- competent oncolytic adenovirus OBP-301 in human esophageal cancer TE4 and TE6 cells with an EMT phenotype. Transforming growth factor-ß (TGF-ß) administration induced the EMT phenotype with spindleshaped morphology, upregulation of mesenchymal markers and EMT transcription factors, migration, and chemoresistance in TE4 and TE6 cells. OBP-301 significantly inhibited the EMT phenotype via E1 accumulation. EMT cancer cells were susceptible to OBP-301 via massive autophagy induction. OBP-301 suppressed tumor growth and lymph node metastasis of TE4 cells co-inoculated with TGF-ß-secreting fibroblasts. Our results suggest that OBP-301 inhibits the TGF-ß-induced EMT phenotype in human esophageal cancer cells. OBP-301-mediated E1A overexpression is a promising antitumor strategy to inhibit EMT-mediated esophageal cancer progression.


Subject(s)
Epithelial-Mesenchymal Transition , Esophageal Neoplasms , Adenoviridae/genetics , Cell Line, Tumor , Epithelial-Mesenchymal Transition/genetics , Esophageal Neoplasms/pathology , Humans , Transforming Growth Factor beta/pharmacology , Transforming Growth Factors
13.
Gan To Kagaku Ryoho ; 49(10): 1127-1129, 2022 Oct.
Article in Japanese | MEDLINE | ID: mdl-36281608

ABSTRACT

Pancreatic cancer has poor prognosis despite the various developed multimodal treatment strategies. Currently, neoadjuvant chemotherapy and immunotherapy have attracted substantial attention as effective treatment strategies. However, amplifying immune response with existing treatments is difficult. We developed telomerase-specific oncolytic adenoviruses (OAs), including OBP-301 that is currently tested in a clinical trial of combined anti-PD-1 antibody and p53-armed OBP- 301 variant(OBP-702). OAs have immune-modulation functions and induce CD8+ T cells into tumors by releasing immunogenic cell death markers, such as extracellular adenosine triphosphate. Here, we investigated the effectiveness of OBP- 702 in pancreatic cancer treatments, focusing on the influence on CD8+ memory T cells.


Subject(s)
Oncolytic Virotherapy , Pancreatic Neoplasms , Telomerase , Humans , Adenosine Triphosphate/metabolism , Adenoviridae/genetics , Adenoviridae/metabolism , CD8-Positive T-Lymphocytes , Cell Line, Tumor , Genes, p53 , Memory T Cells , Pancreatic Neoplasms/genetics , Pancreatic Neoplasms/therapy , Telomerase/genetics , Telomerase/metabolism , Tumor Suppressor Protein p53/genetics , Clinical Trials as Topic , Pancreatic Neoplasms
14.
Int J Cancer ; 149(2): 347-357, 2021 07 15.
Article in English | MEDLINE | ID: mdl-33662150

ABSTRACT

Esophageal cancer is a disease showing poor prognosis. Although combination chemotherapy using cisplatin (CDDP) and 5-fluorouracil is standard for unresectable esophageal cancer, the response rate is 35%. Cancer stem cells (CSCs) and inflammation are reportedly responsible for the poor prognosis of esophageal cancer. However, comprehensive analyses have not been conducted and proposals for progress remain lacking. Iron is known to be a key factor in the stemness of CSCs. Our study focused on the therapeutic potential of iron control using iron chelators for CSCs in esophageal cancer. Among 134 immunohistochemically analyzed cases, Nanog expression was high in 98 cases and low in 36 cases. High Nanog expression correlated with low overall and disease-free survivals. The iron chelators deferasirox (DFX) and SP10 suppressed the proliferation and expression of stemness markers in TE8 and OE33 cells. DFX and SP10 did not induce compensatory interleukin (IL)-6 secretion, although CDDP did result in high induction. Moreover, BBI608 and SSZ, as other CSC-targeting drugs, could not suppress the expression of stemness markers. Overall, Nanog expression appears related to poor prognosis in esophageal cancer patients, and inhibition of stemness and compensatory IL-6 secretion by iron chelators may offer a novel therapeutic strategy for esophageal cancer.


Subject(s)
Drug Resistance, Neoplasm/drug effects , Esophageal Neoplasms/drug therapy , Gene Expression Profiling/methods , Iron Chelating Agents/administration & dosage , Nanog Homeobox Protein/genetics , Nanog Homeobox Protein/metabolism , Animals , Cell Line, Tumor , Cell Proliferation/drug effects , Cell Survival/drug effects , Cisplatin/pharmacology , Esophageal Neoplasms/genetics , Esophageal Neoplasms/metabolism , Female , Gene Expression Regulation, Neoplastic/drug effects , Humans , Iron Chelating Agents/pharmacology , Male , Mice , Nanog Homeobox Protein/drug effects , Neoplastic Stem Cells/drug effects , Neoplastic Stem Cells/metabolism , Prognosis , Sequence Analysis, RNA , Up-Regulation/drug effects , Xenograft Model Antitumor Assays
15.
Cancer Immunol Immunother ; 70(5): 1405-1417, 2021 May.
Article in English | MEDLINE | ID: mdl-33151368

ABSTRACT

Immune checkpoint inhibitors including anti-programmed cell death 1 (PD-1) antibody have recently improved clinical outcome in certain cancer patients; however, osteosarcoma (OS) patients are refractory to PD-1 blockade. Oncolytic virotherapy has emerged as novel immunogenic therapy to augment antitumor immune response. We developed a telomerase-specific replication-competent oncolytic adenovirus OBP-502 that induces lytic cell death via binding to integrins. In this study, we assessed the combined effect of PD-1 blockade and OBP-502 in OS cells. The expression of coxsackie and adenovirus receptor (CAR), integrins αvß3 and αvß5, and programmed cell death ligand 1 (PD-L1) was analyzed in two murine OS cells (K7M2, NHOS). The cytopathic activity of OBP-502 in both cells was analyzed using the XTT assay. OBP-502-induced immunogenic cell death was assessed by analyzing the level of extracellular ATP and high-mobility group box protein B1 (HMGB1). Subcutaneous tumor models for K7M2 and NHOS cells were used to evaluate the antitumor effect and number of tumor-infiltrating CD8+ cells in combination therapy. K7M2 and NHOS cells showed high expression of integrins αvß3 and αvß5, but not CAR. OBP-502 significantly suppressed the viability of both cells, in which PD-L1 expression and the release of ATP and HMGB1 were significantly increased. Intratumoral injection of OBP-502 significantly augmented the efficacy of PD-1 blockade on subcutaneous K2M2 and NHOS tumor models via enhancement of tumor-infiltrating CD8+ T cells. Our results suggest that telomerase-specific oncolytic virotherapy is a promising antitumor strategy to promote the efficacy of PD-1 blockade in OS.


Subject(s)
Antineoplastic Agents/therapeutic use , Immune Checkpoint Inhibitors/therapeutic use , Immunotherapy/methods , Lymphocytes, Tumor-Infiltrating/immunology , Oncolytic Virotherapy/methods , Osteosarcoma/therapy , Skin Neoplasms/therapy , Adenoviridae/genetics , Animals , Cell Line, Tumor , Disease Models, Animal , Female , Humans , Mice , Mice, Inbred BALB C , Programmed Cell Death 1 Receptor/antagonists & inhibitors , Telomerase/genetics
16.
BMC Cancer ; 21(1): 102, 2021 Jan 28.
Article in English | MEDLINE | ID: mdl-33509150

ABSTRACT

BACKGROUND: Peritoneal dissemination often develops in gastric cancer. Tumor-associated macrophages (TAMs) are present in the peritoneal cavity of gastric cancer patients with peritoneal dissemination, facilitating tumor progression. However, the mechanism by which macrophages differentiate into tumor-associated macrophages in the peritoneal cavity is not well understood. In this study, the interplay between gastric cancer-derived extracellular vesicles (EVs) and macrophages was investigated. METHODS: The association between macrophages and EVs in peritoneal ascitic fluid of gastric cancer patients, or from gastric cancer cell lines was examined, and their roles in differentiation of macrophages and potentiation of the malignancy of gastric cancer were further explored. RESULTS: Immunofluorescent assays of the ascitic fluid showed that M2 macrophages were predominant along with the cancer cells in the peritoneal cavity. EVs purified from gastric cancer cells, as well as malignant ascitic fluid, differentiated peripheral blood mononuclear cell-derived macrophages into the M2-like phenotype, which was demonstrated by their morphology and expression of CD163/206. The macrophages differentiated by gastric cancer-derived EVs promoted the migration ability of gastric cancer cells, and the EVs carried STAT3 protein. CONCLUSION: EVs derived from gastric cancer play a role by affecting macrophage phenotypes, suggesting that this may be a part of the underlying mechanism that forms the intraperitoneal cancer microenvironment.


Subject(s)
Extracellular Vesicles/pathology , Leukocytes, Mononuclear/pathology , Macrophages/pathology , Peritoneal Neoplasms/secondary , Stomach Neoplasms/pathology , Animals , Apoptosis , Cell Movement , Cell Proliferation , Extracellular Vesicles/metabolism , Female , Humans , Leukocytes, Mononuclear/metabolism , Macrophage Activation , Macrophages/metabolism , Mice , Mice, Inbred BALB C , Mice, Nude , Peritoneal Neoplasms/metabolism , Stomach Neoplasms/metabolism , Tumor Cells, Cultured , Xenograft Model Antitumor Assays
17.
Mol Ther ; 28(3): 794-804, 2020 03 04.
Article in English | MEDLINE | ID: mdl-31991110

ABSTRACT

The clinical benefit of monotherapy involving immune checkpoint inhibitors (ICIs) such as anti-programmed death-1 antibody (PD-1 Ab) is limited to small populations. We previously developed a telomerase-specific oncolytic adenovirus, Telomelysin (OBP-301), the safety of which was confirmed in a phase I clinical study. Here, we examined the potential of OBP-502, an OBP-301 variant, as an agent for inducing immunogenic cell death (ICD) and synergistically enhancing the efficacy of OBP-502 with PD-1 Ab using CT26 murine colon cancer and PAN02 murine pancreatic cancer cell lines. OBP-502 induced the release of ICD molecules such as adenosine triphosphate (ATP) and high-mobility group box protein 1 (HMGB1) from CT26 and PAN02 cells, leading to recruitment of CD8-positive lymphocytes and inhibition of Foxp3-positive lymphocyte infiltration into tumors. Combination therapy involving OBP-502 intratumoral administration and PD-1 Ab systemic administration significantly suppressed the growth of not only OBP-502-treated tumors but also tumors not treated with OBP-502 (so-called abscopal effect) in CT26 and PAN02 bilateral subcutaneous tumor models, in which active recruitment of CD8-positve lymphocytes was observed even in tumors not treated with OBP-502. This combined efficacy was similar to that observed in a CT26 rectal orthotopic tumor model involving liver metastases. In conclusion, telomerase-specific oncolytic adenoviruses are promising candidates for combined therapies with ICIs.


Subject(s)
Adenoviridae/immunology , Antineoplastic Agents, Immunological/pharmacology , Genetic Therapy , Immunomodulation , Oncolytic Virotherapy , Oncolytic Viruses/immunology , Telomerase/immunology , Adenoviridae/genetics , Animals , Antineoplastic Agents, Immunological/therapeutic use , Cell Line, Tumor , Combined Modality Therapy , Cytotoxicity, Immunologic , Disease Models, Animal , Drug Synergism , Genetic Therapy/adverse effects , Genetic Therapy/methods , Humans , Lymphocytes, Tumor-Infiltrating/immunology , Lymphocytes, Tumor-Infiltrating/metabolism , Lymphocytes, Tumor-Infiltrating/pathology , Mice , Neoplasms/genetics , Neoplasms/immunology , Neoplasms/therapy , Oncolytic Virotherapy/adverse effects , Oncolytic Virotherapy/methods , Oncolytic Viruses/genetics , Programmed Cell Death 1 Receptor/antagonists & inhibitors , T-Lymphocytes/drug effects , T-Lymphocytes/immunology , T-Lymphocytes/metabolism , Xenograft Model Antitumor Assays
18.
Int J Mol Sci ; 22(2)2021 Jan 17.
Article in English | MEDLINE | ID: mdl-33477279

ABSTRACT

Oncolytic virotherapy is one of the most promising, emerging cancer therapeutics. We generated three types of telomerase-specific replication-competent oncolytic adenovirus: OBP-301; a green fluorescent protein (GFP)-expressing adenovirus, OBP-401; and Killer-Red-armed OBP-301. These oncolytic adenoviruses are driven by the human telomerase reverse transcriptase (hTERT) promoter; therefore, they conditionally replicate preferentially in cancer cells. Fluorescence imaging enables visualization of invasion and metastasis in vivo at the subcellular level; including molecular dynamics of cancer cells, resulting in greater precision therapy. In the present review, we focused on fluorescence imaging applications to develop precision targeting for oncolytic virotherapy. Cell-cycle imaging with the fluorescence ubiquitination cell cycle indicator (FUCCI) demonstrated that combination therapy of an oncolytic adenovirus and a cytotoxic agent could precisely target quiescent, chemoresistant cancer stem cells (CSCs) based on decoying the cancer cells to cycle to S-phase by viral treatment, thereby rendering them chemosensitive. Non-invasive fluorescence imaging demonstrated that complete tumor resection with a precise margin, preservation of function, and prevention of distant metastasis, was achieved with fluorescence-guided surgery (FGS) with a GFP-reporter adenovirus. A combination of fluorescence imaging and laser ablation using a KillerRed-protein reporter adenovirus resulted in effective photodynamic cancer therapy (PDT). Thus, imaging technology and the designer oncolytic adenoviruses may have clinical potential for precise cancer targeting by indicating the optimal time for administering therapeutic agents; accurate surgical guidance for complete resection of tumors; and precise targeted cancer-specific photosensitization.


Subject(s)
Neoplasms/therapy , Oncolytic Virotherapy/methods , Oncolytic Virotherapy/trends , Adenoviridae/genetics , Animals , Antineoplastic Agents , Cell Line, Tumor , Fluorescence , Green Fluorescent Proteins/metabolism , Humans , Neoplasms/virology , Oncolytic Viruses/genetics , Optical Imaging/methods , Precision Medicine/methods , Promoter Regions, Genetic/genetics , Telomerase/genetics , Telomerase/metabolism
19.
J Orthop Sci ; 25(3): 503-506, 2020 May.
Article in English | MEDLINE | ID: mdl-31174966

ABSTRACT

BACKGROUND: This study was performed to re-examine the incidence of falls from heights while clearing snow in Akita prefecture after implementation of preventive countermeasures and a public service campaign. METHODS: This was a retrospective analysis of registration data from the section regarding prevention against natural disasters in the Akita prefectural office. The study population comprised all patients who fell from heights while clearing snow and were transported to the emergency department from December 2015 to March 2018. RESULTS: In total, 168 individuals (159 male, 9 female) fell from heights while clearing snow. Their mean age was 65.7 years (range, 19-92 years). The largest proportion of falls occurred in January (54.2%; 91 patients). The most common region in which the falls occurred was the inland part of the southern district of Akita prefecture, which accounted for 73.8% of the falls (124 patients). Individuals aged ≥65 years constituted 52.9% of the patients (n = 89). The most common mechanism of injury was a fall from a rooftop, which accounted for 62.5% of the falls (105 patients), followed by a fall from a ladder (31.0%; 52 patients). The most common sites of injury were the upper and lower extremities (39.2%; 66 patients), followed by the spine (36.3%; 61 patients). Seven of the patients died. CONCLUSIONS: Compared with a previous study by Hatakeyama et al. (Falls from heights while clearing snow in Akita Prefecture. Rinsyou Seikeigeka Clin Orthop Surg 2013 Nov; 48(11): 1091-4 [in Japanese]), the total number of patients who fell from heights while clearing snow decreased by one-half. However, the number of persons aged ≥65 years increased and constituted 52.9% of the patients, reflecting the rapidly expanding older population in Akita prefecture.


Subject(s)
Accidental Falls/prevention & control , Accidental Falls/statistics & numerical data , Health Education/methods , Snow , Adult , Aged , Aged, 80 and over , Emergency Service, Hospital , Female , Humans , Incidence , Japan/epidemiology , Male , Middle Aged , Retrospective Studies
20.
Int J Cancer ; 144(4): 828-840, 2019 02 15.
Article in English | MEDLINE | ID: mdl-30367467

ABSTRACT

Lymph node metastasis is a pathognomonic feature of spreading tumors, and overcoming metastasis is a challenge in attaining more favorable clinical outcomes. Esophageal cancer is an aggressive tumor for which lymph node metastasis is a strong poor prognostic factor, and the tumor microenvironment (TME), and cancer-associated fibroblasts (CAFs) in particular, has been implicated in esophageal cancer progression. CAFs play a central role in the TME and have been reported to provide suitable conditions for the progression of esophageal cancer, similar to their role in other malignancies. However, little is known concerning the relevance of CAFs to the lymph node metastasis of esophageal cancer. Here, we used clinical samples of esophageal cancer to reveal that CAFs promote lymph node metastasis and subsequently verified the intercellular relationships in vitro and in vivo using an orthotopic metastatic mouse model. In the analysis of clinical samples, FAP+ CAFs were strongly associated with lymph node metastasis rather than with other prognostic factors. Furthermore, CAFs affected the ability of esophageal cancer cells to acquire metastatic phenotypes in vitro; this finding was confirmed by data from an in vivo orthotopic metastatic mouse model showing that the number of lymph node metastases increased upon injection of cocultured cancer cells and CAFs. In summary, we verified in vitro and in vivo that the accumulation of CAFs enhances the lymph node metastasis of ESCC. Our data suggest that CAF targeted therapy can reduce lymph node metastasis and improve the prognosis of patients with esophageal cancer in the future.


Subject(s)
Cancer-Associated Fibroblasts/pathology , Carcinoma, Squamous Cell/pathology , Esophageal Neoplasms/pathology , Tumor Microenvironment , Aged , Animals , Cell Line, Tumor , Cell Movement , Female , Humans , Lymphatic Metastasis , Male , Mice, Inbred BALB C , Mice, Nude , Middle Aged , Survival Analysis , Transplantation, Heterologous
SELECTION OF CITATIONS
SEARCH DETAIL