Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 122
Filter
Add more filters

Country/Region as subject
Publication year range
1.
Cancer Cell Int ; 19: 299, 2019.
Article in English | MEDLINE | ID: mdl-31787847

ABSTRACT

BACKGROUND: Photoimmunotherapy (PIT) employing antibody-photosensitizer conjugates is a promising treatment for cancer. However, the fixed antigen specificity severely limits the efficacy and the applicability. Here we describe a universal strategy for PIT of cancer by using a near-infrared (NIR) photosensitizer IRDye700DX-conjugated NeutrAvidin, designated as AvIR, together with various biotinylated antibodies (BioAbs) for cellular targeting. METHODS: Cytotoxicity of AvIR-mediated PIT was evaluated by fluorescence imaging and cell viability assay. Phototoxic effect on tumorigenicity was assessed by tumorsphere-formation assay and Matrigel invasion assay. Cancer stem cell-like side-population (SP) cells were identified by flow cytometry. RESULTS: CHO cells stably expressing carcinoembryonic antigen or EpCAM were pre-labeled with each BioAb for the corresponding antigen, followed by AvIR administration. NIR light irradiation specifically killed the targeted cells, but not off-targets, demonstrating that the AvIR-mediated PIT does work as expected. CSC-like subpopulation of MCF-7 cells (CD24low/CD44high) and SP of HuH-7 cells (CD133+/EpCAM+) were effectively targeted and photokilled by AvIR-PIT with anti-CD44 BioAb or anti-CD133/anti-EpCAM BioAbs, respectively. As results, the neoplastic features of the cell lines were sufficiently suppressed. Cancer-associated fibroblast (CAF)-targeted AvIR-PIT by using anti-fibroblast activation protein BioAb showed an abolishment of CAF-enhanced clonogenicity of MCF-7 cells. CONCLUSIONS: Collectively, our results demonstrate that AvIR-mediated PIT can greatly broaden the applicable range of target specificity, with feasibility of efficacious and integrative control of CSC and its microenvironment.

2.
BMC Cancer ; 17(1): 89, 2017 01 31.
Article in English | MEDLINE | ID: mdl-28143428

ABSTRACT

BACKGROUND: BK-UM (CRM197) is a mutant form of diphtheria toxin and a specific inhibitor of heparin-binding epidermal growth factor-like growth factor (HB-EGF). We assessed the safety, pharmacokinetics, recommended dose, and efficacy of BK-UM in patients with recurrent ovarian cancer (OC) or peritoneal cancer (PC), and measured HB-EGF levels in serum and abdominal fluid after BK-UM administration. METHODS: Eleven patients with advanced or recurrent OC or PC were enrolled and treated with BK-UM via the intraperitoneal route. The dose was escalated (1.0, 2.0, 3.3, and 5.0 mg/m2) using a 3 + 3 design. RESULTS: Eight of 11 patients completed treatment. No dose-limiting toxicity (DLT) was experienced at dose levels 1 (1.0 mg/m2) and 2 (2.0 mg/m2). Grade 3 transient hypotension as an adverse event (defined as a DLT in the present study) was observed in two of four patients at dose level 3 (3.3 mg/m2). Treatment with BK-UM was associated with decreases in HB-EGF levels in serum and abdominal fluid in seven of 11 patients and five of eight patients, respectively. Clinical outcomes included a partial response in one patient, stable disease in five patients, and progressive disease in five patients. CONCLUSIONS: BK-UM was well tolerated at doses of 1.0 and 2.0 mg/m2, with evidence for clinical efficacy in patients with recurrent OC or PC. A dose of 2.0 mg/m2 BK-UM is recommended for subsequent clinical trials. TRIAL REGISTRATION: This trial was prospectively performed as an investigator-initiated clinical trial. The trial numbers are UMIN000001002 and UMIN000001001, with registration dates of 1/30/2008 and 2/4/2008, respectively. UMIN000001001 was registered as a trial for the continuous administration of BK-UM after UMIN000001002 .


Subject(s)
Bacterial Proteins/administration & dosage , Neoplasm Recurrence, Local/drug therapy , Ovarian Neoplasms/drug therapy , Peritoneal Neoplasms/drug therapy , Aged , Bacterial Proteins/pharmacokinetics , Dose-Response Relationship, Drug , Female , Heparin-binding EGF-like Growth Factor/metabolism , Humans , Middle Aged , Neoplasm Recurrence, Local/metabolism , Ovarian Neoplasms/metabolism , Peritoneal Neoplasms/metabolism
3.
Int J Cancer ; 135(11): 2697-710, 2014 Dec 01.
Article in English | MEDLINE | ID: mdl-24740257

ABSTRACT

Conventional photodynamic therapy (PDT) for cancer is limited by the insufficient efficacy and specificity of photosensitizers. We herein describe a highly effective and selective tumor-targeted PDT using a near-infrared (NIR) photosensitizer, IRDye700DX, conjugated to a human monoclonal antibody (Ab) specific for carcinoembryonic antigen (CEA). The antitumor effects of this Ab-assisted PDT, called photoimmunotherapy (PIT), were investigated in vitro and in vivo. The Ab-IRDye conjugate induced potent cytotoxicity against CEA-positive tumor cells after NIR-irradiation, whereas CEA-negative cells were not affected at all, even in the presence of excess photoimmunoconjugate. We found an equivalent phototoxicity and a predominant plasma membrane localization of Ab-IRDye after both one and six hours of incubation. Either no or little caspase activation and membrane peroxidation were observed in PIT-treated cells and a panel of scavengers for reactive oxygen species showed only partial inhibition of the phototoxic effect. Strikingly, Ab-IRDye retained significant phototoxicity even under hypoxia. We established a xenograft model, which allowed us to sensitively investigate the therapeutic efficacy of PIT by non-invasive bioluminescence imaging. Luciferase-expressing MKN-45-luc human gastric carcinoma cells were subcutaneously implanted into both flanks of nude mice. NIR-irradiation was performed for only the tumor on one side. In vivo imaging and measurement of the tumor size revealed that a single PIT treatment, with intraperitoneal administration of Ab-IRDye and subsequent NIR-irradiation, caused rapid cell death and significant inhibition of tumor growth, but only on the irradiated side. Together, these data suggest that Ab-IRDye-mediated PIT has great potential as an anticancer therapeutics targeting CEA-positive tumors.


Subject(s)
Antibodies, Monoclonal/therapeutic use , Carcinoembryonic Antigen/immunology , Immunotherapy , Neoplasms/drug therapy , Photochemotherapy , Photosensitizing Agents/therapeutic use , Animals , Antibodies, Monoclonal/immunology , Apoptosis/drug effects , Blotting, Western , Carcinoembryonic Antigen/metabolism , Cell Proliferation/drug effects , Female , Flow Cytometry , Fluorescent Dyes/therapeutic use , Humans , Immunoconjugates/administration & dosage , Lipid Peroxidation , Mice , Mice, Inbred BALB C , Mice, Nude , Neoplasms/immunology , Neoplasms/metabolism , Tumor Cells, Cultured , Xenograft Model Antitumor Assays
4.
Mol Ther ; 21(3): 561-9, 2013 Mar.
Article in English | MEDLINE | ID: mdl-23070115

ABSTRACT

Glioblastoma multiforme (GBM) remains an untreatable human brain malignancy. Despite promising preclinical studies using oncolytic herpes simplex virus (oHSV) vectors, efficacy in patients has been limited by inefficient virus replication in tumor cells. This disappointing outcome can be attributed in part to attenuating mutations engineered into these viruses to prevent replication in normal cells. Alternatively, retargeting of fully replication-competent HSV to tumor-associated receptors has the potential to achieve tumor specificity without impairment of oncolytic activity. Here, we report the establishment of an HSV retargeting system that relies on the combination of two engineered viral glycoproteins, gD and gB, to mediate highly efficient HSV infection exclusively through recognition of the abundantly expressed epidermal growth factor receptor (EGFR) on glioblastoma cells. We demonstrate efficacy in vitro and in a heterotopic tumor model in mice. Evidence for systemically administered virus homing to the tumor mass is presented. Treatment of orthotopic primary human GBM xenografts demonstrated prolonged survival with up to 73% of animals showing a complete response as confirmed by magnetic resonance imaging. Our study describes an approach to HSV retargeting that is effective in a glioma model and may be applicable to the treatment of a broad range of tumor types.


Subject(s)
ErbB Receptors/metabolism , Glioblastoma/therapy , Oncolytic Virotherapy/methods , Simplexvirus/genetics , Animals , Cell Line, Tumor , Chlorocebus aethiops , Cricetinae , Female , Genetic Vectors , HT29 Cells , Humans , Magnetic Resonance Imaging , Mice , Mice, Inbred BALB C , Mice, Nude , Plasmids , Recombination, Genetic , Simplexvirus/physiology , Treatment Outcome , Vero Cells , Virus Replication , Xenograft Model Antitumor Assays
5.
Circ J ; 77(11): 2651-63, 2013.
Article in English | MEDLINE | ID: mdl-24107398

ABSTRACT

High-density lipoprotein (HDL) and low-density lipoprotein (LDL) particles transport cholesterol in plasma and play an important role in cellular cholesterol homeostasis, which influences cell function. The risk of coronary artery disease (CAD) associated with high levels of LDL-cholesterol (LDL-C) can be reduced by treatment with statins, which reduce LDL-C levels by inhibiting cellular cholesterol synthesis. However, patients who are treated with high doses of statins, especially secondary CAD prevention, regardless of their resulting LDL-C levels, are still at high risk of CAD. Therefore, there has been growing interest in HDL-directed therapies. Inhibitors of cholesteryl ester transfer protein (CETP) substantially increase HDL-C levels (by 31-138%). However, it is still unclear whether or not CETP inhibitors can reduce the risk of CAD associated with low HDL-C levels, while reconstituted HDL or apolipoprotein A-I mimetic peptides increase the functionality of HDL. Low levels of HDL-C are often complicated with metabolic disorders, including hypertriglyceridemia, metabolic syndrome, and type 2 diabetes mellitus, and lifestyle changes are effective for correcting these conditions. Physical activity and exercise training increase HDL-C levels, especially HDL2-C levels, by multiple mechanisms. Therefore, although using HDL-directed therapies that increase HDL-C levels and/or improve the function of HDL is a reasonable approach for reducing the residual risk of CAD as a complement to LDL-C-lowering therapy, lifestyle modifications including exercise to improve metabolic disorders should be considered as the first option.


Subject(s)
Cholesterol, HDL/blood , Coronary Artery Disease , Diabetes Mellitus, Type 2 , Exercise , Hydroxymethylglutaryl-CoA Reductase Inhibitors/therapeutic use , Hypertriglyceridemia , Metabolic Syndrome , Apolipoprotein A-I/blood , Cholesterol/blood , Cholesterol, LDL/blood , Coronary Artery Disease/blood , Coronary Artery Disease/therapy , Diabetes Mellitus, Type 2/blood , Diabetes Mellitus, Type 2/therapy , Female , Humans , Hypertriglyceridemia/blood , Hypertriglyceridemia/therapy , Male , Metabolic Syndrome/blood , Metabolic Syndrome/therapy
6.
Proc Natl Acad Sci U S A ; 107(32): 14199-204, 2010 Aug 10.
Article in English | MEDLINE | ID: mdl-20660741

ABSTRACT

TAL1 plays pivotal roles in vascular and hematopoietic developments through the complex with LMO2 and GATA1. Hemangioblasts, which have a differentiation potential for both endothelial and hematopoietic lineages, arise in the primitive streak and migrate into the yolk sac to form blood islands, where primitive hematopoiesis occurs. ZFAT (a zinc-finger gene in autoimmune thyroid disease susceptibility region/an immune-related transcriptional regulator containing 18 C(2)H(2)-type zinc-finger domains and one AT-hook) was originally identified as an immune-related transcriptional regulator containing 18 C(2)H(2)-type zinc-finger domains and one AT-hook, and is highly conserved among species. ZFAT is thought to be a critical transcription factor involved in immune-regulation and apoptosis; however, developmental roles for ZFAT remain unknown. Here we show that Zfat-deficient (Zfat(-/-)) mice are embryonic-lethal, with impaired differentiation of hematopoietic progenitor cells in blood islands, where ZFAT is exactly expressed. Expression levels of Tal1, Lmo2, and Gata1 in Zfat(-/-) yolk sacs are much reduced compared with those of wild-type mice, and ChIP-PCR analysis revealed that ZFAT binds promoter regions for these genes in vivo. Furthermore, profound reduction in TAL1, LMO2, and GATA1 protein expressions are observed in Zfat(-/-) blood islands. Taken together, these results suggest that ZFAT is indispensable for mouse embryonic development and functions as a critical transcription factor for primitive hematopoiesis through direct-regulation of Tal1, Lmo2, and Gata1. Elucidation of ZFAT functions in hematopoiesis might lead to a better understanding of transcriptional networks in differentiation and cellular programs of hematopoietic lineage and provide useful information for applied medicine in stem cell therapy.


Subject(s)
Cell Differentiation/genetics , Hematopoiesis/genetics , Hematopoietic Stem Cells/cytology , Transcription Factors/physiology , Animals , Basic Helix-Loop-Helix Transcription Factors , Embryo, Mammalian , Gene Expression Regulation, Developmental , Mice , Mice, Knockout , Proto-Oncogene Proteins , T-Cell Acute Lymphocytic Leukemia Protein 1 , Transcription Factors/genetics , Yolk Sac/blood supply , Zinc Fingers/genetics
7.
Mol Cancer ; 11: 46, 2012 Jul 25.
Article in English | MEDLINE | ID: mdl-22830422

ABSTRACT

BACKGROUND: We previously established a three-dimensional (3-D) colonic crypt model using HKe3 cells which are human colorectal cancer (CRC) HCT116 cells with a disruption in oncogenic KRAS, and revealed the crucial roles of oncogenic KRAS both in inhibition of apoptosis and in disruption of cell polarity; however, the molecular mechanism of KRAS-induced these 3-D specific biological changes remains to be elucidated. RESULTS: Among the genes that were upregulated by oncogenic KRAS in this model, we focused on the phosphodiesterase 4B (PDE4B) of which expression levels were found to be higher in clinical tumor samples from CRC patients in comparison to those from healthy control in the public datasets of gene expression analysis. PDE4B2 was specifically overexpressed among other PDE4 isoforms, and re-expression of oncogenic KRAS in HKe3 cells resulted in PDE4B overexpression. Furthermore, the inhibition of PDE4 catalytic activity using rolipram reverted the disorganization of HCT116 cells into the normal physiologic state of the epithelial cell polarity by inducing the apical assembly of ZO-1 (a tight junction marker) and E-cadherin (an adherens junction marker) and by increasing the activity of caspase-3 (an apoptosis marker) in luminal cavities. Notably, rolipram reduced the AKT phosphorylation, which is known to be associated with the disruption of luminal cavity formation and CRC development. Similar results were also obtained using PDE4B2-shRNAs. In addition, increased expression of PDE4B mRNA was found to be correlated with relapsed CRC in a public datasets of gene expression analysis. CONCLUSIONS: These results collectively suggested that PDE4B is upregulated by oncogenic KRAS, and also that the inhibition of PDE4 catalytic activity can induce both epithelial cell polarity and luminal apoptosis in CRC, thus highlighting the utility of our 3-D culture (3 DC) model for the KRAS-induced development of CRC in 3-D microenvironment. Indeed, using this model, we found that PDE4B is a promising candidate for a therapeutic target as well as prognostic molecular marker in CRC. Further elucidation of the signaling network of PDE4B2 in 3 DC would provide a better understanding of CRC in vivo.


Subject(s)
Apoptosis/genetics , Colorectal Neoplasms/genetics , Colorectal Neoplasms/metabolism , Cyclic Nucleotide Phosphodiesterases, Type 4/genetics , Proto-Oncogene Proteins c-akt/metabolism , Apoptosis/drug effects , Cell Line, Tumor , Cluster Analysis , Cyclic Nucleotide Phosphodiesterases, Type 4/metabolism , Enzyme Activation/drug effects , Gene Expression Profiling , Gene Expression Regulation, Neoplastic/drug effects , Genes, ras , HCT116 Cells , Humans , Phosphodiesterase 4 Inhibitors/pharmacology , Phosphorylation/drug effects , RNA Interference , Recurrence , Rolipram/pharmacology , Spheroids, Cellular , Tight Junctions/metabolism , Tumor Cells, Cultured
8.
Biochem Biophys Res Commun ; 425(1): 107-12, 2012 Aug 17.
Article in English | MEDLINE | ID: mdl-22828507

ABSTRACT

ZFAT, originally identified as a candidate susceptibility gene for autoimmune thyroid disease, has been reported to be involved in apoptosis, development and primitive hematopoiesis. Zfat is highly expressed in T- and B-cells in the lymphoid tissues, however, its physiological function in the immune system remains totally unknown. Here, we generated the T cell-specific Zfat-deficient mice and demonstrated that Zfat-deficiency leads to a remarkable reduction in the number of the peripheral T cells. Intriguingly, a reduced expression of IL-7Rα and the impaired responsiveness to IL-7 for the survival were observed in the Zfat-deficient T cells. Furthermore, a severe defect in proliferation and increased apoptosis in the Zfat-deficient T cells following T cell receptor (TCR) stimulation was observed with a reduced IL-2Rα expression as well as a reduced IL-2 production. Thus, our findings reveal that Zfat is a critical regulator in peripheral T cell homeostasis and its TCR-mediated response.


Subject(s)
CD4-Positive T-Lymphocytes/immunology , Homeostasis/immunology , Receptors, Antigen, T-Cell/immunology , Transcription Factors/physiology , Animals , Apoptosis/immunology , CD4 Lymphocyte Count , Cell Proliferation , Interleukin-7/pharmacology , Mice , Mice, Mutant Strains , Receptors, Interleukin-7/biosynthesis , Transcription Factors/genetics
9.
Stem Cells ; 29(10): 1485-95, 2011 Oct.
Article in English | MEDLINE | ID: mdl-21919130

ABSTRACT

Cancer stem cells (CSCs) that display tumor-initiating properties have recently been identified. CD133, a surface glycoprotein linked to organ-specific stem cells, has been described as a marker of CSCs in different tumor types. We herein identify and characterize CSCs in human uterine carcinosarcoma (malignant mixed Müllerian tumor), which is one of the most aggressive and therapy-resistant gynecological malignancies and is considered to be of mesodermal origin. The CD133(+) population was increased in uterine carcinosarcoma, and this population showed biphasic properties in the primary tumor. CD133(+) cells predominantly formed spheres in culture and were able to differentiate into mesenchymal lineages. CD133(+) cells were more resistant to cisplatin/paclitaxel-induced cytotoxicity in comparison with CD133(-) cells. A real-time polymerase chain reaction analysis of the genes implicated in stem cell maintenance revealed that CD133(+) cells express significantly higher levels of Oct4, Nanog, Sox2, and Bmi1 than CD133(-) cells. Moreover, CD133(+) cells showed a high expression level of Pax2 and Wnt4, which are genes essential for Müllerian duct formation. These CD133(+) cells form serially transplantable tumors in vivo and the resulting CD133(+) tumors replicated the EpCAM, vimentin, and estrogen and progesterone receptor expression of the parent tumor, indicating that CSCs likely differentiated into cells comprising the uterine carcinosarcoma tissue. Moreover, strong CD133 expression in both epithelial and mesenchymal elements in primary tumor demonstrated significant prognostic value. These findings suggest that CD133(+) cells have the characteristics of CSCs and Müllerian mesenchymal progenitors.


Subject(s)
Antigens, CD/metabolism , Carcinosarcoma/pathology , Gene Expression Regulation, Neoplastic , Glycoproteins/metabolism , Neoplastic Stem Cells/pathology , Peptides/metabolism , Uterine Neoplasms/pathology , AC133 Antigen , Animals , Antigens, Neoplasm/metabolism , Carcinosarcoma/genetics , Carcinosarcoma/metabolism , Cell Adhesion Molecules/metabolism , Cell Differentiation , Cell Line, Tumor , Cell Separation/methods , Cell Transformation, Neoplastic/metabolism , Cell Transformation, Neoplastic/pathology , Cisplatin/pharmacology , Drug Resistance, Neoplasm , Epithelial Cell Adhesion Molecule , Estrogen Receptor beta/metabolism , Female , Flow Cytometry , Homeodomain Proteins/metabolism , Humans , Immunohistochemistry , Mesenchymal Stem Cells/cytology , Mesenchymal Stem Cells/metabolism , Mice , Mice, Inbred BALB C , Mice, Nude , Mixed Tumor, Mullerian/metabolism , Mixed Tumor, Mullerian/pathology , Nanog Homeobox Protein , Neoplastic Stem Cells/cytology , Neoplastic Stem Cells/drug effects , Octamer Transcription Factor-3/metabolism , PAX2 Transcription Factor/metabolism , Paclitaxel/pharmacology , SOXB1 Transcription Factors/metabolism , Uterine Neoplasms/genetics , Uterine Neoplasms/metabolism , Vimentin/metabolism , Wnt4 Protein/metabolism
10.
J Biomed Biotechnol ; 2012: 853879, 2012.
Article in English | MEDLINE | ID: mdl-22547929

ABSTRACT

The transduction of T cells to express chimeric T-cell antigen receptor (CAR) is an attractive strategy for adaptive immunotherapy for cancer, because the CAR can redirect the recognition specificity of T cells to tumor-associated antigens (TAAs) on the surface of target cells, thereby avoiding the limitations of HLA restriction. However, there are considerable problems with the clinical application of CAR, mostly due to its xenogeneic components, which could be immunogenic in humans. Moreover, while extensive studies on the CARs have been performed, the detailed molecular mechanisms underlying the activation of CAR-grafted T cells remain unclear. In order to eliminate potential immunogenicity and investigate the molecular basis of the CAR-mediated T-cell activation, we constructed a novel CAR (CAR57-28ζ) specific for one of the most important TAAs, epithelial cell adhesion molecule (EpCAM), using only human-derived genes. We revealed that in Jurkat T cells, lentivirally expressed CAR57-28ζ can transmit the T-cell-activating signals sufficient to induce IL-2 production upon EpCAM stimulation. An immunofluorescent analysis clearly showed that the CAR57-28ζ induces the formation of signaling clusters containing endogenous CD3ζ at the CAR/EpCAM interaction interface. These results suggest that this CAR gene may be safely and effectively applied for adaptive T-cell immunotherapy.


Subject(s)
Antigens, Neoplasm/metabolism , Cell Adhesion Molecules/metabolism , Receptors, Antigen, T-Cell/metabolism , Recombinant Fusion Proteins/metabolism , Single-Chain Antibodies/metabolism , Amino Acid Sequence , Antigens, Neoplasm/chemistry , Antigens, Neoplasm/immunology , Base Sequence , Cell Adhesion Molecules/chemistry , Cell Adhesion Molecules/immunology , Cloning, Molecular/methods , Epithelial Cell Adhesion Molecule , Humans , Interleukin-2/immunology , Interleukin-2/metabolism , Jurkat Cells , Molecular Sequence Data , Phosphorylation , Receptors, Antigen, T-Cell/chemistry , Receptors, Antigen, T-Cell/genetics , Receptors, Antigen, T-Cell/immunology , Recombinant Fusion Proteins/chemistry , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/immunology , Single-Chain Antibodies/chemistry , Single-Chain Antibodies/genetics , Single-Chain Antibodies/immunology
11.
Circ J ; 76(9): 2241-8, 2012.
Article in English | MEDLINE | ID: mdl-22785328

ABSTRACT

BACKGROUND: The aim of the present study was to compare 2 direct measurements for low-density lipoprotein cholesterol (LDL-C) with the Friedewald calculation (LDL-C [F]) in serum and their relationship with size-and charge-based LDL subfractions in serum ultracentrifugation fractions in patients with hypercholesterolemia (HC). METHODS AND RESULTS: Serum samples from 283 HC patients who participated in a statin trial (the PATROL trial) were analyzed. Homogeneous assays for LDL-C were performed using reagents from Sekisui Medical (LDL-C [Se]) and Kyowa Medex (LDL-C [Ky]). Charge-based LDL subfractions were analyzed by capillary isotachophoresis (cITP). In whole serum in HC patients at baseline, LDL-C (Se) and LDL-C (Ky) negatively and positively deviated, respectively, from LDL-C (F). The negative deviation of LDL-C (Se) from LDL-C (F) increased with increasing LDL-C, while the positive deviation of LDL-C (Ky) from LDL-C (F) was positively correlated with charge-modified LDL (cmLDL) as analyzed by cITP. In serum d>1.006 g/ml and >1.040 g/ml fractions (LDL and small, dense LDL fractions, respectively), the deviation of LDL-C (Ky) from LDL-C (Se) was positively correlated with LDL-apoB (the number of LDL particles) and cmLDL. CONCLUSIONS: The 2 homogenous assays for LDL-C differed with regard to reactivity toward LDL particles and cmLDL in patients with HC. Direct measurement of LDL-C that reflects modified LDL, could be a better marker for the risk of coronary heart disease.


Subject(s)
Cholesterol, LDL/blood , Coronary Disease/blood , Hypercholesterolemia/blood , Apolipoproteins B/blood , Biomarkers/blood , Female , Humans , Male , Ultracentrifugation
12.
Mol Ther ; 19(3): 507-14, 2011 Mar.
Article in English | MEDLINE | ID: mdl-20924362

ABSTRACT

The safety and efficacy of viral therapies for solid tumors can be enhanced by redirecting the virus infection to tumor-specific cell-surface markers. Successful retargeting of herpes simplex virus type 1 (HSV-1) has been achieved using vectors that carry a modified envelope glycoprotein D (gD) engineered to interact directly with novel receptors. In addition, soluble bridging molecules (adapters) have been used to link gD indirectly to cell-specific receptors. Here, we describe the development of an adapter connecting gD to the common tumor antigen carcinoembryonic antigen (CEA). The adapter consisted of a CEA-specific single-chain antibody fused to the gD-binding region of the gD receptor, herpes virus entry mediator (HVEM). We used this adapter in combination with a vector that is detargeted for recognition of the widely expressed gD receptor nectin-1, but retains an intact binding region for the less common HVEM. We show that the adapter enabled infection of HSV-resistant Chinese hamster ovary (CHO) cells expressing ectopic CEA and nectin-1/CEA-bearing human gastric carcinoma cells that are resistant to the vector alone. We observed cell-to-cell spread following adapter-mediated infection in vitro and reduced tumor growth in vivo, indicating that this method of vector retargeting may provide a novel strategy for tumor-specific delivery of tumoricidal HSV.


Subject(s)
Carcinoembryonic Antigen , Genetic Vectors/genetics , Herpesvirus 1, Human/genetics , Receptors, Tumor Necrosis Factor, Member 14 , Single-Chain Antibodies , Adenocarcinoma/metabolism , Adenocarcinoma/virology , Animals , Antibody Specificity/genetics , Antibody Specificity/immunology , CHO Cells , Carcinoembryonic Antigen/immunology , Carcinoembryonic Antigen/metabolism , Cell Adhesion Molecules/metabolism , Cell Line, Tumor , Cell Proliferation/drug effects , Cricetinae , Cricetulus , Gene Order , Genetic Vectors/administration & dosage , HEK293 Cells , Humans , Male , Mice , Mice, Inbred BALB C , Mice, Nude , Nectins , Receptors, Tumor Necrosis Factor, Member 14/genetics , Receptors, Tumor Necrosis Factor, Member 14/metabolism , Single-Chain Antibodies/genetics , Single-Chain Antibodies/metabolism , Stomach Neoplasms/metabolism , Stomach Neoplasms/virology
13.
Chemotherapy ; 58(2): 110-7, 2012.
Article in English | MEDLINE | ID: mdl-22507917

ABSTRACT

BACKGROUND: Carcinoembryonic antigen (CEA) is frequently overexpressed in various types of human cancers and is associated with cell adhesion. There are three possible mechanisms of cancer therapy that employ anti-CEA antibody (Ab): Ab-dependent cell-mediated cytotoxicity (ADCC), complement-dependent cytotoxicity (CDC) or the prevention of CEA interaction with the extracellular matrix and/or intercellular adhesion molecules resulting in anoikis. In this study, the effect of C2-74, a human anti-CEA monoclonal Ab was evaluated. METHODS: ADCC, CDC and anoikis assays in combination with C2-74 and an anticancer drug (5-fluorouracil or cisplatin) were investigated using tumor cell lines (MKN-45, MKN-74 and KATO III). In the anoikis assay, other human anti-CEA Abs and mouse anti-CEA-related cell adhesion molecule 6 Abs were also investigated using HLC-1 cells. RESULTS: Additive cytotoxicity was observed when the anticancer drug and C2-74 on tumor cells were combined in the CDC assays, whereas in the anoikis assay, no such additive effect was observed. Anti-CEA-related cell adhesion molecule 6 Abs, but not anti-CEA Abs, accelerated anoikis in HLC-1 cells. CONCLUSION: A mechanism for the additive antitumor effect when an anticancer drug and C2-74 are combined is indicated mainly by CDC activity but is irrelevant to anoikis in tumor cells.


Subject(s)
Antibodies, Monoclonal/pharmacology , Antineoplastic Agents/pharmacology , Carcinoembryonic Antigen/immunology , Anoikis/drug effects , Antibodies, Monoclonal/immunology , Cell Adhesion Molecules/immunology , Cell Line, Tumor , Cisplatin/pharmacology , Complement C2/immunology , Fluorouracil/pharmacology , Humans
14.
Biochem Biophys Res Commun ; 408(2): 282-6, 2011 May 06.
Article in English | MEDLINE | ID: mdl-21501587

ABSTRACT

KRAS-induced actin-interacting protein (KRAP) was originally characterized as a filamentous-actin-interacting protein. We have recently found that KRAP is an associated molecule with inositol 1,4,5-trisphosphate receptor (IP(3)R) and is critical for the proper subcellular localization and function of IP(3)R. However, the molecular mechanisms underlying the regulation of IP(3)R by KRAP remain elusive. In this report, to determine the critical region of KRAP protein for the regulation of IP(3)R, we generate several mutants of KRAP and examine the association with IP(3)R using coimmunoprecipitation and confocal imaging assays. Coimmunoprecipitations using the deletion mutants reveal that amino-acid residues 1-218 but not 1-199 of KRAP interact with IP(3)R, indicating that the 19-length amino-acid residues (200-218) are essential for the association with IP(3)R. This critical region is highly conserved between human and mouse KRAP. Within the critical region, substitutions of two phenylalanine residues (Phe202/Phe203) in mouse KRAP to alanines result in failure of the association with IP(3)R, suggesting that the two consecutive phenylalanine residues are indispensable for the association. Moreover, the KRAP-knockdown stable HeLa cells exhibit the inappropriate subcellular localization of IP(3)R, in which exogenous expression of full-length of KRAP properly restores the subcellular localization of IP(3)R, but not the 1-218 or 1-236 mutant, indicating that the residual carboxyl-terminal region is also required for the proper subcellular localization of KRAP-IP(3)R complex. All these results provide insight into the understandings for the molecular mechanisms underlying the regulation of IP(3)R, and would reveal a potent strategy for the drug development targeting on IP(3)R.


Subject(s)
Inositol 1,4,5-Trisphosphate Receptors/metabolism , Membrane Proteins/metabolism , Microfilament Proteins/metabolism , Amino Acid Sequence , Animals , HeLa Cells , Humans , Immunoprecipitation , Inositol 1,4,5-Trisphosphate Receptors/chemistry , Inositol 1,4,5-Trisphosphate Receptors/genetics , Membrane Proteins/chemistry , Membrane Proteins/genetics , Mice , Microfilament Proteins/chemistry , Microfilament Proteins/genetics , Molecular Sequence Data , Phenylalanine/chemistry , Phenylalanine/genetics
15.
Biochem Biophys Res Commun ; 408(2): 214-7, 2011 May 06.
Article in English | MEDLINE | ID: mdl-21457704

ABSTRACT

KRAS-induced actin-interacting protein (KRAP) was originally characterized as a filamentous- actin-interacting protein. We have recently found that KRAP is an associated molecule with inositol 1,4,5-trisphosphate (IP(3)) receptor (IP(3)R) and is responsible for the proper subcellular localization of IP(3)R. Since it remains unknown whether KRAP regulates the IP(3)R-mediated Ca(2+) signaling, we herein examined the effects of KRAP on the IP(3)R-mediated Ca(2+) release by Ca(2+) imagings in the cultured HEK293 or MCF7 cells. Reduction of KRAP protein by KRAP-specific siRNA diminishes ATP-induced Ca(2+) release and the ATP-induced Ca(2+) release is completely quenched by the pretreatment with the IP(3)R inhibitor but not with the ryanodine receptor inhibitor, indicating that KRAP regulates IP(3)R-mediated Ca(2+) release. To further reveal mechanistic insights into the regulation of IP(3)R-mediated Ca(2+) release by KRAP, we examined the effects of the KRAP-knockdown on the releasable Ca(2+) content of intracellular Ca(2+) stores. Consequently, reduction of KRAP does not affect the amount of ionophore- or Ca(2+)-ATPase inhibitor-induced Ca(2+) release in the HEK293 cells, indicating that releasable Ca(2+) content of intracellular Ca(2+) stores is not altered by KRAP. Thus, KRAP is involved in the proper regulation of IP(3)R-mediated Ca(2+) release.


Subject(s)
Calcium/metabolism , Inositol 1,4,5-Trisphosphate Receptors/metabolism , Membrane Proteins/metabolism , Microfilament Proteins/metabolism , Animals , Cell Line, Tumor , HEK293 Cells , Humans , Membrane Proteins/genetics , Mice , Mice, Knockout , Microfilament Proteins/genetics
16.
Biochem Biophys Res Commun ; 407(2): 438-43, 2011 Apr 08.
Article in English | MEDLINE | ID: mdl-21420385

ABSTRACT

Three inositol 1,4,5-trisphosphate receptor (IP(3)R) subtypes are differentially expressed among tissues and function as the Ca(2+) release channel on specialized endoplasmic reticulum (ER) membranes. The proper subcellular localization of IP(3)R is crucial for its proper function, but this molecular mechanism is unclear. KRAS-induced actin-interacting protein (KRAP) was originally identified as a cancer-related molecule, and is involved in the regulation of whole-body energy homeostasis and pancreatic exocrine system. We herein identified IP(3)R as an associated molecule with KRAP in vivo, and the association was validated by the co-immunoprecipitation and confocal immunostaining studies in mouse tissues including liver and pancreas. The association of KRAP with IP(3)R was also observed in the human epithelial cell lines including HCT116, HeLa and HEK293 cells. Intriguingly, KRAP interacts with distinct subtypes of IP(3)R in a tissue-dependent manner, i.e. IP(3)R1 and IP(3)R2 in the liver and IP(3)R2 and IP(3)R3 in the pancreas. The NH(2)-terminal amino acid residues 1-610 of IP(3)R are critical for the association with KRAP and KRAP-IP(3)R complex resides in a specialized ER but not a typical reticular ER. Furthermore, the localization of particular IP(3)R subtypes in tissues from KRAP-deficient mice is obviously disturbed, i.e. IP(3)R1 and IP(3)R2 in the liver and IP(3)R2 and IP(3)R3 in the pancreas. These findings demonstrate that KRAP physically associates with IP(3)R and regulates the proper localization of IP(3)R in the epithelial cells in vivo and cultured cells, and might shed light on the Ca(2+) signaling underlying physiological cellular programs, cancer development and metabolism-related diseases.


Subject(s)
Epithelial Cells/metabolism , Inositol 1,4,5-Trisphosphate Receptors/metabolism , Microfilament Proteins/metabolism , Animals , Calcium/metabolism , Calcium Signaling , Dogs , Endoplasmic Reticulum/metabolism , HEK293 Cells , HeLa Cells , Humans , Immunoprecipitation , Liver/metabolism , Mice , Mice, Knockout , Microfilament Proteins/genetics , Pancreas/metabolism , Tissue Distribution
17.
Curr Opin Obstet Gynecol ; 23(1): 24-30, 2011 Feb.
Article in English | MEDLINE | ID: mdl-20966750

ABSTRACT

PURPOSE OF REVIEW: Therapeutics targeting the ErbB protein family receptors have not always yielded favorable or successful results in present cancer therapy. This review discusses the possibility of the clinical adaptation of targeting against heparin-binding epidermal growth factor-like growth factor (HB-EGF), one of the ligands of the ErbB system, in ovarian cancer therapy. RECENT FINDINGS: We have previously described the results of studies concerning roles of HB-EGF in tumor formation in ovarian cancer. In brief, lisophosphatidic acid (LPA) and HB-EGF are predominantly expressed in advanced ovarian cancer, and LPA-induced, a disintegrin and metalloprotease-mediated ectodomain shedding of HB-EGF was found to be critical to tumor formation. We also noted that exogenous expression of HB-EGF enhanced tumor formation but inhibition blocked both extracellular signal-related kinase and serine/threonine protein kinase activation. Finally we investigated the antitumor effects of CRM197 - a specific HB-EGF inhibitor - on ovarian cancer cells by evaluating human ovarian cancer cell proliferation. SUMMARY: We discuss alternative strategies to develop the chemotherapeutic agent based on targeting ErbB family ligands rather than their receptors. A phase I study of CRM197 for advanced ovarian cancer has already begun, which is the first approved trial of ErbB-ligand-targeted therapy. We also discuss clinical adaptations based on combination of CRM197 with other conventional chemotherapeutic agents.


Subject(s)
Epidermal Growth Factor/antagonists & inhibitors , Intercellular Signaling Peptides and Proteins/metabolism , Ovarian Neoplasms/drug therapy , Ovarian Neoplasms/metabolism , Antineoplastic Agents/therapeutic use , Bacterial Proteins/therapeutic use , Cell Proliferation/drug effects , Clinical Trials, Phase I as Topic , Female , Heparin-binding EGF-like Growth Factor , Humans , Lysophospholipids/metabolism , Ovarian Neoplasms/pathology , Protein Serine-Threonine Kinases/metabolism , Signal Transduction
18.
Cell Mol Biol Lett ; 16(1): 89-100, 2011 Mar.
Article in English | MEDLINE | ID: mdl-21225468

ABSTRACT

ZFAT was originally identified as an immune-related transcriptional regulator containing 18 C2H2-type zinc-finger domains and one AT-hook. ZFAT is highly conserved among species and functions as an anti-apoptotic molecule in the lymphoblastic leukemia cell line, MOLT-4. We recently demonstrated that ZFAT is an essential molecule for hematopoietic differentiation in blood islands through the direct regulation of particular transcriptional factors, including Tal1, for endothelial cell assembly, and for the branch point formation of capillary-like structures. However, the molecular mechanisms underlying the anti-apoptotic function of ZFAT remain unknown. Here, we report that ZFAT knockdown by small interfering RNA induced apoptosis in mouse embryonic fibroblasts (MEFs). This response had been similarly observed for MOLT-4 cells. To explore the molecular mechanisms for ZFAT in anti-apoptotic function in both MEFs and MOLT-4 cells, microarray expression analysis and quantitative RT-PCR were done. Of interest was that Bcl-2 and Il6st were identified as commonly down-regulated genes by the depletion of ZFAT for both MEFs and MOLT-4 cells. These results suggest that ZFAT is a critical molecule for cell survival in MEFs and MOLT-4 cells at least in part through the regulation of the apoptosis involved in the BCL-2- and IL6st-mediated pathways. Further elucidation of the molecular functions for ZFAT might shed light on the cellular programs in the mesoderm-derived cells.


Subject(s)
Fibroblasts/metabolism , Transcription Factors/physiology , Animals , Apoptosis , Cell Survival , Cytokine Receptor gp130/genetics , Cytokine Receptor gp130/metabolism , Down-Regulation , Mice , Oligonucleotide Array Sequence Analysis , Proto-Oncogene Proteins c-bcl-2/genetics , Proto-Oncogene Proteins c-bcl-2/metabolism , RNA Interference , RNA, Small Interfering/metabolism , Transcription Factors/genetics , Transcription Factors/metabolism
19.
Int J Cancer ; 127(11): 2707-17, 2010 Dec 01.
Article in English | MEDLINE | ID: mdl-20499311

ABSTRACT

A number of therapeutic strategies targeting epidermal growth factor receptor (EGFR) have not always led to success in the present state of breast cancer therapy. Notably, there is currently no way to treat trastuzumab-resistant and triple-negative breast cancer (TNBC). Here, we found that heparin-binding epidermal growth factor-like growth factor (HB-EGF), a member of the EGFR ligands, was predominantly expressed in breast cancer and that treatment with crossreacting material 197 (CRM197), a specific inhibitor of HB-EGF, blocked ERK as well as AKT activation via complexes of EGFR and unknown growth factor receptors in TNBC or through complexes of EGFR and human epidermal growth factor receptor-2 in trastuzumab-resistant breast cancer, caused significant cell apoptosis and inhibited tumor growth. Accordingly, we can provide a novel concept that a certain EGFR ligand is recognized as a rational target against breast cancer. In addition, it is plausible that CRM197 could be an effective anticancer agent for molecularly targeted therapies.


Subject(s)
Antibodies, Monoclonal/pharmacology , Breast Neoplasms/drug therapy , Breast Neoplasms/metabolism , Intercellular Signaling Peptides and Proteins/metabolism , Animals , Antibodies, Monoclonal, Humanized , Antineoplastic Agents/pharmacology , Bacterial Proteins/pharmacology , Breast Neoplasms/enzymology , Cell Line, Tumor , Cell Transformation, Neoplastic/metabolism , Cetuximab , Drug Delivery Systems/methods , Drug Resistance, Neoplasm , ErbB Receptors/metabolism , Heparin-binding EGF-like Growth Factor , Humans , Intercellular Signaling Peptides and Proteins/biosynthesis , Mice , Mice, Inbred NOD , Mice, SCID , Oncogene Protein v-akt/metabolism , Receptor, ErbB-2/metabolism , Receptors, Estrogen/metabolism , Receptors, Progesterone/metabolism , Reproducibility of Results , Signal Transduction , Trastuzumab
20.
Cancer Sci ; 101(11): 2351-60, 2010 Nov.
Article in English | MEDLINE | ID: mdl-20726858

ABSTRACT

Pancreatic cancer is one of the most lethal malignancies. Epidermal growth factor receptor (EGFR), HER3, Akt, and amphiregulin have been recognized as targets for pancreatic cancer therapy. Although gemcitabine + erlotinib has been the recommended chemotherapy for pancreatic cancer, the prognosis is extremely poor. The development of molecularly targeted therapies has been required for patients with pancreatic cancer. To assess the validation of amphiregulin as a target for pancreatic cancer therapy, we examined its expression in pancreatic cancer using real-time PCR analyses and ELISA. We also measured the apoptotic cell rate using TUNEL assays. In addition, alterations in signaling pathways were detected by immunoblotting analyses. Treatment with gemcitabine, which reduced the cell viability and augmented the cell apoptotic rate, activated and subsequently attenuated ERK and EGFR signals. However, gemcitabine, paclitaxel, or cisplatin treatment enhanced the Akt activation, heterodimer formation of EGFR with HER3, and secretion of amphiregulin, indicating that the presence of gemcitabine promoted the activity of targeted molecules including amphiregulin, Akt, and HER3 for pancreatic cancer therapy. Combined treatment with an inhibitor for amphiregulin and gemcitabine, paclitaxel, or cisplatin induced synergistic antitumor effects, accompanied by the suppression of Akt and ERK activation. Blockade of amphiregulin suppressed the activities of EGFR, HER3, and Akt and the expression of amphiregulin itself. According to this evidence, combination chemotherapy of conventional anticancer drugs plus an inhibitor for amphiregulin would allow us to provide more favorable clinical outcomes for patients with pancreatic cancer.


Subject(s)
ErbB Receptors/metabolism , Extracellular Signal-Regulated MAP Kinases/metabolism , Glycoproteins/metabolism , Intercellular Signaling Peptides and Proteins/metabolism , Pancreatic Neoplasms/metabolism , Proto-Oncogene Proteins c-akt/metabolism , Receptor, ErbB-3/metabolism , Amphiregulin , Antibodies/immunology , Antibodies/pharmacology , Apoptosis/drug effects , Cell Line, Tumor , Cell Survival/drug effects , Cisplatin/pharmacology , Deoxycytidine/analogs & derivatives , Deoxycytidine/pharmacology , Dose-Response Relationship, Drug , Drug Synergism , EGF Family of Proteins , Enzyme Activation , Enzyme-Linked Immunosorbent Assay , ErbB Receptors/antagonists & inhibitors , ErbB Receptors/genetics , Glycoproteins/genetics , Glycoproteins/immunology , Humans , Immunoblotting , Intercellular Signaling Peptides and Proteins/genetics , Intercellular Signaling Peptides and Proteins/immunology , Paclitaxel/pharmacology , Pancreatic Neoplasms/genetics , Pancreatic Neoplasms/pathology , Proto-Oncogene Proteins c-akt/genetics , Receptor, ErbB-3/genetics , Reverse Transcriptase Polymerase Chain Reaction , Signal Transduction/drug effects , Gemcitabine
SELECTION OF CITATIONS
SEARCH DETAIL