Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 10 de 10
Filter
1.
Cancer Sci ; 110(11): 3595-3602, 2019 Nov.
Article in English | MEDLINE | ID: mdl-31512325

ABSTRACT

Coxsackievirus and adenovirus receptor (CAR) is a single-pass transmembrane protein that is associated with adenoviral infection. CAR is involved in the formation of epithelial tight junctions and promotes tumor growth in some cancers. Previously, we developed mouse monoclonal antibodies against human CAR and found that one, mu6G10A, significantly inhibited tumor growth in xenografts of human cancer cells. Herein, we generated and characterized a mouse-human chimeric anti-CAR antibody (ch6G10A) from mu6G10A. ch6G10A had binding activity, inducing antibody-dependent cellular cytotoxicity and complement-dependent cytotoxicity, and in vivo anti-tumor activity against CAR-expressing prostate cancer DU-145 cells. In addition, cancer tissue array analysis confirmed that CAR is highly expressed in neuroendocrine lung cancers including small cell lung cancer, and treatment with ch6G10A effectively inhibited in vivo subcutaneous tumor growth of NCI-H69 small cell lung cancer cells in nude mice. Moreover, treatment with mu6G10A effectively inhibited both in vivo orthotopic tumor growth and distant metastatic formation in mouse xenograft models of a highly metastatic subline of human small cell lung cancer DMS273 cells. These results suggest that targeting therapy to CAR with a therapeutic antibody might be effective against several cancer types including small cell lung cancer.


Subject(s)
Antibodies, Monoclonal/therapeutic use , Coxsackie and Adenovirus Receptor-Like Membrane Protein/antagonists & inhibitors , Lung Neoplasms/therapy , Prostatic Neoplasms/therapy , Small Cell Lung Carcinoma/therapy , Animals , Antibody-Dependent Cell Cytotoxicity , Complement System Proteins/immunology , Coxsackie and Adenovirus Receptor-Like Membrane Protein/immunology , Coxsackie and Adenovirus Receptor-Like Membrane Protein/metabolism , Endocrine Gland Neoplasms/metabolism , Endocrine Gland Neoplasms/therapy , Female , Humans , Lung Neoplasms/metabolism , Male , Mice , Mice, Inbred BALB C , Mice, Nude , Molecular Targeted Therapy , Prostatic Neoplasms/metabolism , Small Cell Lung Carcinoma/metabolism , Xenograft Model Antitumor Assays
2.
PLoS Pathog ; 15(6): e1007817, 2019 06.
Article in English | MEDLINE | ID: mdl-31163079

ABSTRACT

Schistosome infection persists for decades. Parasites are in close contact with host peripheral blood immune cells, yet little is known about the regulatory interactions between parasites and these immune cells. Here, we report that extracellular vesicles (EVs) released from Schistosoma japonicum are taken up primarily by macrophages and other host peripheral blood immune cells and their miRNA cargo transferred into recipient cells. Uptake of S. japonicum EV miR-125b and bantam miRNAs into host cells increased macrophage proliferation and TNF-α production by regulating the corresponding targets including Pros1, Fam212b, and Clmp. Mice infected with S. japonicum exhibit an increased population of monocytes and elevated levels of TNF-α. Reduction of host monocytes and TNF-α level in S. japonicum infected mice led to a significant reduction in worm and egg burden and pathology. Overall, we demonstrate that S. japonicum EV miRNAs can regulate host macrophages illustrating parasite modulation of the host immune response to facilitate parasite survival. Our findings provide valuable insights into the schistosome-host interaction which may help to develop novel intervention strategies against schistosomiasis.


Subject(s)
Extracellular Vesicles/immunology , Macrophages/immunology , MicroRNAs/immunology , RNA, Helminth/immunology , Schistosoma japonicum/immunology , Animals , Calcium-Binding Proteins , Carrier Proteins/immunology , Coxsackie and Adenovirus Receptor-Like Membrane Protein/immunology , Macrophages/parasitology , Mice , Monocytes/immunology , Monocytes/parasitology , RAW 264.7 Cells , Rabbits , Tumor Necrosis Factor-alpha/immunology
3.
Sci Rep ; 7(1): 14177, 2017 10 26.
Article in English | MEDLINE | ID: mdl-29074882

ABSTRACT

Oncolytic virotherapy has the disadvantage of being unsuitable for systemic delivery due to immune elimination. Liposomal encapsulation is well-recognized to reduce immune elimination and enhance the stability of drugs in the bloodstream. In the present study, the potential of liposome-encapsulated plasmid DNA of telomerase-specific oncolytic adenovirus (TelomeScan) expressing GFP (Lipo-pTS) as an oncolytic adenoviral agent suitable for systemic delivery was investigated. Lipo-pTS, which has a diameter of 40-50 nm, showed potent antitumor effects on HCT116 colon carcinoma cells in vitro and in vivo. Tumor selectivity of Lipo-pTS was independent of coxsackie and adenovirus receptor (CAR). Importantly, Lipo-pTS reduced production of adenovirus-neutralizing antibodies (AdNAbs) after intravenous administration into immune-competent mice compared to TelomeScan, and even in the presence of AdNAbs, Lipo-pTS maintained strong cytotoxicity. In conclusion, Lipo-pTS has the potential to become an oncolytic adenoviral agent suitable for systemic delivery with the characteristics of CAR-independent antitumor activity and a stealth effect on the immune system.


Subject(s)
Adenoviridae/genetics , Antibodies, Neutralizing/immunology , DNA, Viral/administration & dosage , Liposomes/chemistry , Oncolytic Viruses/genetics , Adenoviridae/immunology , Animals , Coxsackie and Adenovirus Receptor-Like Membrane Protein/immunology , Coxsackie and Adenovirus Receptor-Like Membrane Protein/metabolism , DNA, Viral/chemistry , DNA, Viral/immunology , Dose-Response Relationship, Immunologic , Female , HCT116 Cells , Humans , Liposomes/administration & dosage , Liposomes/pharmacology , Mice, Inbred BALB C , Oncolytic Virotherapy/methods , Oncolytic Viruses/immunology , Plasmids/administration & dosage , Plasmids/genetics , Telomerase/genetics , Xenograft Model Antitumor Assays
4.
Mol Immunol ; 68(2 Pt A): 234-43, 2015 Dec.
Article in English | MEDLINE | ID: mdl-26391350

ABSTRACT

Effective use of adenovirus-5 (Ad5) in cancer therapy is heavily dependent on the degree to which the virus's natural tropism can be subverted to one that favours tumour cells. This is normally achieved through either engineering of the viral fiber knob or the use of bispecific adaptors that display both adenovirus and tumour antigen receptors. One of the main limitations of these strategies is the need to tailor each engineering event to any given tumour antigen. Here, we explore bispecific adaptors that can utilise established anti-cancer therapeutic antibodies. Conjugates containing bacterially derived antibody binding motifs are efficient at retargeting virus to antibody targets. Here, we develop a humanized strategy whereby we synthesise a re-targeting adaptor based on a chimeric Ad5 ligand/antibody receptor construct. This adaptor acts as a molecular bridge analogous to therapeutic antibody mediated cross-linking of cytotoxic effector and tumour cells during immunotherapy. As a proof or principle, we demonstrate how this adaptor allows efficient viral recognition and entry into carcinoma cells through the therapeutic monoclonal antibodies Herceptin/trastuzumab and bavituximab. We show that targeting can be augmented by use of contemporary antibody enhancement strategies such as the selective elimination of competing serum IgG using "receptor refocusing" enzymes and we envisage that further improvements are achievable by enhancing the affinities between the adaptor and its ligands. Humanized bispecific adaptors offer the promise of a versatile retargeting technology that can exploit both clinically approved adenovirus and therapeutic antibodies.


Subject(s)
Adenoviridae/genetics , Antibodies, Bispecific/immunology , Antibodies, Monoclonal/immunology , Antigens, Neoplasm/immunology , Antigens, Viral/immunology , Receptors, IgG/immunology , Trastuzumab/immunology , Adenoviridae/immunology , Amino Acid Sequence , Antibodies, Bispecific/chemistry , Antibodies, Bispecific/genetics , Antibodies, Monoclonal/chemistry , Antigens, Neoplasm/chemistry , Antigens, Neoplasm/genetics , Antigens, Viral/chemistry , Antigens, Viral/genetics , Antineoplastic Agents/chemistry , Antineoplastic Agents/immunology , Bacterial Proteins/chemistry , Bacterial Proteins/metabolism , Breast Neoplasms/genetics , Breast Neoplasms/immunology , Breast Neoplasms/pathology , Breast Neoplasms/therapy , Cell Line, Tumor , Coxsackie and Adenovirus Receptor-Like Membrane Protein/genetics , Coxsackie and Adenovirus Receptor-Like Membrane Protein/immunology , Female , Genetic Vectors , Glycoside Hydrolases/chemistry , Glycoside Hydrolases/metabolism , HEK293 Cells , Humans , Immunoconjugates/chemistry , Immunoconjugates/genetics , Immunotherapy/methods , Molecular Sequence Data , Protein Binding , Protein Engineering , Receptor, ErbB-2/genetics , Receptor, ErbB-2/immunology , Receptors, IgG/chemistry , Receptors, IgG/genetics , Recombinant Fusion Proteins/chemistry , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/immunology , Trastuzumab/chemistry
5.
Kardiologiia ; 54(5): 8-15, 2014.
Article in Russian | MEDLINE | ID: mdl-25177881

ABSTRACT

In 22 patients with heart failure and/or ventricular arrhythmias presumably of inflammatory etiology the results of clinical and instrumental investigation were analyzed and compared to the endomyocardial biopsy data. In the subgroup of patients with left bundle branch block (LBBB) we revealed features indicative of lesser contribution of inflammatory destruction in pathogenesis of cardiomyopathy. The only virus, detected in biopsy samples, was parvovirus B19. Its persistence in myocardium was not related to activity of inflammation and severity of clinical course. Increased expression of Coxsackie adenovirus receptor (CAR) was found in 20 patients. It was not related to inflammatory cells infiltration and virus persistence in myocardium. Patients with most prominent CAR expression were characteried by right heart dilatation, more severe heart failure and absence of LBBB. Enhancement of CAR expression could reflect the attempt of organism to repair intercellular communications between cardiomyocites and to protect cells from the products of necrotic lysis during long standing inflammation.


Subject(s)
Arrhythmias, Cardiac , Coxsackie and Adenovirus Receptor-Like Membrane Protein/immunology , Heart Conduction System/abnormalities , Heart Failure , Heart Ventricles , Myocarditis , Adult , Arrhythmias, Cardiac/diagnosis , Arrhythmias, Cardiac/etiology , Arrhythmias, Cardiac/immunology , Arrhythmias, Cardiac/physiopathology , Biopsy , Brugada Syndrome , Bundle-Branch Block/immunology , Bundle-Branch Block/physiopathology , Cardiac Conduction System Disease , Female , Heart Conduction System/immunology , Heart Conduction System/physiopathology , Heart Failure/diagnosis , Heart Failure/etiology , Heart Failure/physiopathology , Heart Function Tests/methods , Heart Ventricles/pathology , Heart Ventricles/physiopathology , Humans , Male , Middle Aged , Monitoring, Immunologic , Myocarditis/complications , Myocarditis/immunology , Myocarditis/pathology , Prognosis , Risk Assessment , Severity of Illness Index , Statistics as Topic
6.
Xi Bao Yu Fen Zi Mian Yi Xue Za Zhi ; 30(5): 476-9, 484, 2014 May.
Article in Chinese | MEDLINE | ID: mdl-24796741

ABSTRACT

OBJECTIVE: To construct a prokaryotic expression plasmid for CT40L, express the target protein in E. coli, purity the CT40L fusion protein and verify its antigenicity. METHODS: Gene sequences of Coxsackie and adenovirus receptor (CAR), bacteriophage T4 fibritin and mouse CD40L were found out in GenBank. Then functional domains of three molecules were linked to form a fusion sequence which was then optimized for prokaryotic expression. The optimized sequence was cloned into prokaryotic expression vector pET42a(+) to construct the recombinant expression vector pET42a-CT40L. The recombinant vector was transformed into BL21 (DE3) and the fusion protein CT40L/GST was induced by IPTG. The fusion protein was then subjected to purification using GST affinity chromatography and to identification of the immune activity using Western blotting and ELISA. RESULTS: The recombinant expression vector was verified correct by double digestion with Nco I and EcoR I. After IPTG induction, SDS-PAGE showed that the relative molecular mass of the fusion protein was about 78 kDa and that the purity of the purified protein reached 90%. Western blotting and ELISA demonstrated that the purified fusion protein had a valid antigenicity. CONCLUSION: The prokaryotic expression plasmid pET42a-Ct40L was successfully constructed and expressed in E. coli, and the purified fusion protein was proved to have a good antigenicity.


Subject(s)
CD40 Ligand/metabolism , Coxsackie and Adenovirus Receptor-Like Membrane Protein/metabolism , Dendritic Cells/metabolism , Recombinant Fusion Proteins/metabolism , Viral Proteins/metabolism , Adenoviridae/genetics , Adenoviridae/immunology , Adenoviridae/physiology , Amino Acid Sequence , Animals , Blotting, Western , CD40 Antigens/immunology , CD40 Antigens/metabolism , CD40 Ligand/genetics , CD40 Ligand/immunology , Coxsackie and Adenovirus Receptor-Like Membrane Protein/genetics , Coxsackie and Adenovirus Receptor-Like Membrane Protein/immunology , Dendritic Cells/immunology , Dendritic Cells/virology , Enzyme-Linked Immunosorbent Assay , Escherichia coli/genetics , Gene Expression/immunology , Host-Pathogen Interactions/immunology , Mice , Molecular Sequence Data , Protein Binding/immunology , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/immunology , Viral Proteins/genetics , Viral Proteins/immunology
7.
J Leukoc Biol ; 95(6): 867-74, 2014 Jun.
Article in English | MEDLINE | ID: mdl-24659789

ABSTRACT

The cellular and molecular principles that furnish the foundation for ACT of melanoma and their implications for further clinical research are reviewed. The parallel advances in basic immunology, preclinical animal studies, and clinical trials over the last two decades have been integrated successfully with improvements in technology to produce an effective ACT strategy for patients with melanoma. From the initial observation that tumors could be treated effectively by the transfer of immune cells to current strategies using preconditioning with myeloablative therapy before adoptive transfer of native or genetically altered T cells, the role of preclinical animal models is discussed. The importance of the pmel transgenic mouse model in the determination of the mechanisms of lymphodepletion, the ongoing work to identify the optimal T cells for adoptive immunotherapy, and the early impact of the emerging discipline of synthetic biology are highlighted. The clinical consequences of the research described herein are reviewed in the companion manuscript.


Subject(s)
Immunotherapy, Adoptive , Melanoma/therapy , T-Lymphocytes/immunology , Animals , Coxsackie and Adenovirus Receptor-Like Membrane Protein/immunology , Humans , Interleukin-2/pharmacology , Killer Cells, Lymphokine-Activated/immunology , Lymphocyte Depletion , Protein Engineering , Receptors, Antigen, T-Cell/immunology
8.
Tumour Biol ; 34(1): 17-24, 2013 Feb.
Article in English | MEDLINE | ID: mdl-23307165

ABSTRACT

The aim of this study is to elucidate the relation between expression of coxsackie and adenovirus receptor (CAR) and formation of lung cancer. We investigated the expression of CAR by immunohistochemistry, Western blot and real-time RT-PCR in 120 lung cancers. We found that CAR expression in tumor tissues was significantly higher than that in normal lung tissues. CAR expression had a correlation with the histological grade of lung squamous cell carcinoma; however, there was no relationship between the CAR expression and the other clinical pathological features. In vitro, silencing or overexpression of CAR could significantly inhibit or promote colony formation, cell adhesion, and invasion in A549 cells. Our findings demonstrated that CAR may play an essential role in the formation of lung cancer.


Subject(s)
Coxsackie and Adenovirus Receptor-Like Membrane Protein/metabolism , Lung Neoplasms/metabolism , Adult , Aged , Carcinoma, Squamous Cell/genetics , Carcinoma, Squamous Cell/metabolism , Carcinoma, Squamous Cell/pathology , Cell Adhesion , Cell Line, Tumor , Coxsackie and Adenovirus Receptor-Like Membrane Protein/genetics , Coxsackie and Adenovirus Receptor-Like Membrane Protein/immunology , Female , Gene Expression Regulation, Neoplastic , Humans , Lung/metabolism , Lung Neoplasms/genetics , Lung Neoplasms/pathology , Male , Middle Aged , Neoplasm Grading , Neoplasm Invasiveness , RNA Interference , RNA, Small Interfering
9.
Microbes Infect ; 15(1): 18-27, 2013 Jan.
Article in English | MEDLINE | ID: mdl-23108316

ABSTRACT

Coxsackievirus B4 (CV-B4), in presence of antibodies and through a specific viral receptor CAR and Fcγ receptors II and III, can infect monocytes which results in interferon-α synthesis. The antibody-dependent enhancement of CV-B4 infection in the human monocytic-like THP-1 cell line has been investigated. The preincubation of CV-B4 with human plasma or human polyvalent immunoglobulins enhanced the infection of phorbol-myristate-acetate (PMA)-activated THP-1 cell cultures. CV-B4 replicated in these cells as demonstrated by the intracellular detection of infectious particles, viral protein VP1 (immunofluorescence), positive and negative viral RNA (RT-PCR). The viability of infected and control cell cultures was not different up to 20 days post-infection. Activated cell cultures inoculated with CV-B4 harbored intracellular RNA up to 14 days post-infection and produced IFNα that was detected by intracellular immunofluorescence staining as soon as 4 h post-infection with a maximum at 48 h post-infection and by RT-PCR all along the experiment. Together, these data demonstrate that PMA-activated THP-1 cells can be infected with CV-B4, can produce IFNα as a result of interactions between the virus, antibodies and specific receptors. This cellular model can be used to investigate further the mechanism and the result of the antibody-dependent enhancement of CV-B4 infection.


Subject(s)
Enterovirus B, Human/immunology , Immunoglobulins/pharmacology , Monocytes/immunology , Monocytes/virology , Cells, Cultured , Coxsackie and Adenovirus Receptor-Like Membrane Protein/immunology , Coxsackievirus Infections/immunology , Coxsackievirus Infections/virology , Enterovirus B, Human/pathogenicity , Host-Pathogen Interactions/drug effects , Humans , Interferon-alpha/biosynthesis , Interferon-alpha/immunology , Intracellular Space/metabolism , Intracellular Space/virology , Monocytes/drug effects , Monocytes/metabolism , Receptors, IgG/immunology , T-Lymphocytes, Helper-Inducer/immunology , Tetradecanoylphorbol Acetate/analogs & derivatives , Tetradecanoylphorbol Acetate/pharmacology
10.
J Virol ; 86(18): 9590-8, 2012 Sep.
Article in English | MEDLINE | ID: mdl-22787208

ABSTRACT

Adenovirus (Ad) vaccine vectors have proven highly immunogenic in multiple experimental models, but the innate immune responses induced by these vectors remain poorly characterized. Here we report innate cytokine responses to 5 different Ad vectors in 26 rhesus monkeys. Vaccination with adenovirus serotype 35 (Ad35), Ad26, and Ad48 induced substantially higher levels of antiviral (gamma interferon [IFN-γ], 10-kDa gamma interferon-induced protein [IP-10]) and proinflammatory (interleukin 1 receptor antagonist [IL-1RA], IL-6) cytokines than vaccination with Ad5 on day 1 following immunization. In vitro studies with capsid chimeric vectors and receptor-blocking monoclonal antibodies suggested that fiber-receptor interactions, as well as other capsid components, were critical for triggering these innate responses. Moreover, multiple cell populations, including dendritic cells, monocytes/macrophages, and T lymphocytes, contributed to these innate cytokine profiles. These data demonstrate that Ad35, Ad26, and Ad48, which utilize CD46 as their primary cellular receptor, induce significantly greater innate cytokine responses than Ad5, which uses the coxsackievirus and adenovirus receptor (CAR). These differences in innate triggering result in markedly different immunologic milieus for the subsequent generation of adaptive immune responses by these vaccine vectors.


Subject(s)
Adenoviruses, Human/classification , Adenoviruses, Human/immunology , Cytokines/biosynthesis , Immunity, Innate , Macaca mulatta/immunology , Macaca mulatta/virology , Adenoviruses, Human/genetics , Animals , Chemokines/biosynthesis , Chemokines/blood , Coxsackie and Adenovirus Receptor-Like Membrane Protein/immunology , Cytokines/blood , Genetic Vectors/immunology , Humans , In Vitro Techniques , Membrane Cofactor Protein/immunology , Receptors, Virus/immunology , Serotyping , Species Specificity , Vaccination
SELECTION OF CITATIONS
SEARCH DETAIL
...