Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 9 de 9
Filter
Add more filters










Database
Language
Publication year range
1.
J Immunol Methods ; 448: 85-90, 2017 09.
Article in English | MEDLINE | ID: mdl-28614699

ABSTRACT

One major unmet need is improving the sensitivity of immune-diagnostic assays. This is particularly important in the field of biomarker discoveries and monitoring. We have established a novel signal amplification probe system enabling a highly sensitive target detection platform to be used in immuno-assays. The probe consists of a double stranded DNA that can carry a large number of signaling elements such as biotin or fluorescent molecules. The DNA probe anchors to the recognition unit, whether an antibody or an aptamer, by covalent conjugation or by a simple and rapid molecular association process. Binding curves obtained by using the DNA amplification probe are dose dependent and linear over a wide range of antigen concentration. The optimal slopes are characterized by high signals and low background increasing the assay sensitivity and reducing the limit of detection by up to 10-fold compared to biotinylated antibodies commonly used in ELISA systems. When using aptamers in combination with the amplification probe for antigen recognition, the limit of detection is comparable to that obtained by biotinylated antibodies. Biotin labeled aptamers practically cannot be used for detection of low target levels. The DNA amplification probe system enables to expand the range of diagnostic assays including clinical samples and meet research needs.


Subject(s)
Antibodies/isolation & purification , Aptamers, Nucleotide/metabolism , DNA Probes/metabolism , Enzyme-Linked Immunosorbent Assay/methods , Animals , Antibodies/immunology , Antibodies/metabolism , Antibody Specificity , Aptamers, Nucleotide/genetics , Biotinylation , DNA Probes/genetics , Humans , Immunoglobulin G/immunology , Immunoglobulin G/metabolism , Limit of Detection , Mice , Protein Binding , Proto-Oncogene Proteins c-sis/immunology , Proto-Oncogene Proteins c-sis/metabolism , Reproducibility of Results , Thrombin/immunology , Thrombin/metabolism , Tumor Necrosis Factor-alpha/immunology , Tumor Necrosis Factor-alpha/metabolism
2.
FASEB J ; 21(13): 3522-33, 2007 Nov.
Article in English | MEDLINE | ID: mdl-17575264

ABSTRACT

A femtosecond laser beam gene transduction (SG-LBGT) system is described as a novel and efficient method of intradermal (i.d.) nonviral gene delivery in mice by permeabilizing cells utilizing femtosecond laser pulses. Using this approach, significant gene expression and efficient dermal transduction lasting for >7 months were obtained. The ability of this new DNA gene transfer method to enhance genetic vaccination was tested in BALB/C mice. A single i.d. injection of a plasmid (10 microg) containing the hepatitis B virus (HBV) surface antigen (HBsAg), followed by pulses of laser, induced high titers of HBsAg-specific antibodies lasting for >210 days and increased levels of IgG1, IgG2a, IFNgamma, and IL-4, indicating the activation of both Th1 and Th2 cells. Moreover, mice vaccinated using the SG-LBGT followed by challenge with pHBV showed increased protection against viral challenge, as detected by decreased levels of HBV DNA, suggesting an efficient Th1 effect against HBV-infected replicating cells. Tumor growth retardation was induced in vaccinated mice challenged with an HBsAg-expressing syngeneic tumor. In most of the parameters tested, administration of plasmid followed by laser application was significantly more effective and prolonged than that of plasmid alone. Tissue damage was not detected and integration of the plasmid into the host genomic DNA probably did not occur. We suggest that the LBGT method is an efficient and safe technology for in vivo gene expression and vaccination and emphasizes its potential therapeutic applications for i.d. nonviral gene delivery.


Subject(s)
DNA/administration & dosage , Gene Expression , Vaccines, DNA/administration & dosage , Animals , Cells, Cultured , Hepatitis B Surface Antigens/genetics , Hepatitis B Surface Antigens/immunology , Hepatitis B virus/immunology , Immunoglobulin G/metabolism , Interferon-gamma/metabolism , Interleukin-4/metabolism , Lasers , Mice , Mice, Inbred BALB C , Th1 Cells/immunology , Th1 Cells/metabolism , Th2 Cells/immunology , Th2 Cells/metabolism
3.
J Hepatol ; 46(1): 37-44, 2007 Jan.
Article in English | MEDLINE | ID: mdl-17112624

ABSTRACT

BACKGROUND/AIMS: HCV-AB68, a human monoclonal antibody against the envelope protein of hepatitis C virus (HCV), neutralizes HCV in cell-culture and in the HCV-Trimera mouse model. A Phase 1 clinical trial was designed to test safety, tolerability, and antiviral activity of HCV-AB68 in patients with chronic HCV-infection. METHODS/RESULTS: Single doses of HCV-AB68, 0.25-40 mg, administered to 15 patients were well tolerated with no moderate or serious adverse events (SAEs) reported. In six patients, HCV-RNA levels transiently decreased by 2- to 100-fold immediately following infusion and rebound to baseline in 24-48 h. Multiple doses of HCV-AB68, 10-120 mg, were administered to 25 patients. Doses were given weekly for 3 weeks, then 3x a week during the fourth week, after which patients were followed for 3 months. No drug-related SAEs were reported and no specific pattern of adverse events was evident. Eight out of 25 patients had at least a 1-log reduction and 17 had at least a 0.75-log reduction in HCV-RNA levels from baseline at one or more time points following HCV-AB68 infusion. CONCLUSIONS: These data support the investigation of HCV-AB68 in the prevention of recurrent HCV-infection in patients who had received hepatic allografts for end-stage liver disease.


Subject(s)
Antibodies, Monoclonal/therapeutic use , Antiviral Agents/therapeutic use , Hepacivirus/immunology , Hepatitis C Antibodies/therapeutic use , Hepatitis C, Chronic/therapy , Adolescent , Adult , Aged , Animals , Antibodies, Monoclonal/adverse effects , Antiviral Agents/adverse effects , Base Sequence , DNA Primers/genetics , Drug Tolerance , Female , Hepacivirus/genetics , Hepatitis C Antibodies/adverse effects , Hepatitis C, Chronic/virology , Humans , Male , Mice , Middle Aged , RNA, Viral/blood , RNA, Viral/genetics , Safety
4.
Hepatol Res ; 34(4): 228-37, 2006 Apr.
Article in English | MEDLINE | ID: mdl-16520091

ABSTRACT

A simple reproducible and versatile small animal model for hepatitis B virus (HBV) infection is still unavailable. We have generated a simple transient liver-targeted transgenic mouse. Hydrodynamics tail vein injection of a head-to-tail dimer of adw HBV genome (pHBVadwHTD) into immunocompetent mice generated HBsAg and HBeAg expression in both serum and hepatocytes, followed by seroconversion. The injection of pHBVadwHTD into SCID mice generated prolonged HBsAg and HBeAg antigenemia and HBV viremia. Our results demonstrate that hydrodynamic injection of naked DNA could support the generation of HBV particles. We used this model for the assessment of anti-viral agents. Administration of our human monoclonal antibodies, HBV-Ab17(XTL) and HBV-Ab19(XTL), as well as Lamivudine (3TC) treatment suppressed HBV viremia. The model presented herein supports long and stable expression of HBV and will enable determination of various biological questions related to HBV life cycle, mutants and could enhance the development of anti-viral reagents.

5.
J Virol ; 80(6): 2654-64, 2006 Mar.
Article in English | MEDLINE | ID: mdl-16501075

ABSTRACT

Passive immunotherapy is potentially effective in preventing reinfection of liver grafts in hepatitis C virus (HCV)-associated liver transplant patients. A combination of monoclonal antibodies directed against different epitopes may be advantageous against a highly mutating virus such as HCV. Two human monoclonal antibodies (HumAbs) against the E2 envelope protein of HCV were developed and tested for the ability to neutralize the virus and prevent human liver infection. These antibodies, designated HCV-AB 68 and HCV-AB 65, recognize different conformational epitopes on E2. They were characterized in vitro biochemically and functionally. Both HumAbs are immunoglobulin G1 and have affinity constants to recombinant E2 constructs in the range of 10(-10) M. They are able to immunoprecipitate HCV particles from infected patients' sera from diverse genotypes and to stain HCV-infected human liver tissue. Both antibodies can fix complement and form immune complexes, but they do not activate complement-dependent or antibody-dependent cytotoxicity. Upon complement fixation, the monoclonal antibodies induce phagocytosis of the immune complexes by neutrophils, suggesting that the mechanism of viral clearance includes endocytosis. In vivo, in the HCV-Trimera model, both HumAbs were capable of inhibiting HCV infection of human liver fragments and of reducing the mean viral load in HCV-positive animals. The demonstrated neutralizing activities of HCV-AB 68 and HCV-AB 65 suggest that they have the potential to prevent reinfection in liver transplant patients and to serve as prophylactic treatment in postexposure events.


Subject(s)
Antibodies, Monoclonal/therapeutic use , Hepacivirus/immunology , Hepatitis C Antibodies/therapeutic use , Hepatitis C/prevention & control , Liver Transplantation/adverse effects , Viral Envelope Proteins/immunology , Amino Acid Sequence , Animals , Drug Evaluation, Preclinical , Humans , Mice , Molecular Sequence Data , Neutralization Tests , Recurrence , Sequence Analysis, DNA
6.
J Med Virol ; 73(4): 529-35, 2004 Aug.
Article in English | MEDLINE | ID: mdl-15221896

ABSTRACT

Cryptic hepatitis C virus (HCV) infection relates to patients infected chronically with HCV that are seronegative but have HCV-RNA. These patients are not identified by the standard serological tests for HCV, which are based on detection of antibodies to core, NS3 and NS5 antigens. They will, therefore, be wrongly diagnosed as non-infected, and are considered as a potential risk for others. Cryptic HCV infection in dialysis units occurs frequently and, due to medical procedures, is a major factor for contracting the virus when unrecognised. This study was conducted in order to assess the humoral immune responses to E2-antigen in sera of patients infected chronically with HCV. Recombinant E2 protein in enzyme linked immunosorbent assay (ELISA) and Western blot (WB) were used to test the occurrence of anti-E2 antibodies in the sera of patients from the liver clinic and of dialysis patients. The presence of E2 antibodies was found to be correlated with the presence of HCV-RNA and with viral load. Antibodies to the E2 protein could be detected in as many as 30% of the sera from dialysis patients with cryptic HCV infection (HCV-RNA only). The results suggest that detection of anti-E2 antibodies may enhance significantly HCV serological standard testing; especially among patients on dialysis, and that antibodies to envelope E2 protein appear to depend on and correlate with the presence of HCV particles.


Subject(s)
Hepacivirus/immunology , Hepatitis C Antibodies/blood , Hepatitis C, Chronic/virology , Renal Dialysis , Viral Envelope Proteins/immunology , Viremia/virology , Blotting, Western , Enzyme-Linked Immunosorbent Assay , Hepatitis C Antigens/genetics , Hepatitis C Antigens/immunology , Hepatitis C, Chronic/immunology , Humans , RNA, Viral/blood , Recombinant Proteins/immunology , Viral Envelope Proteins/genetics , Viral Load , Viremia/immunology
7.
Mol Ther ; 8(5): 769-76, 2003 Nov.
Article in English | MEDLINE | ID: mdl-14599810

ABSTRACT

Current therapies for chronic hepatitis B virus (HBV) infection are limited in their effect on viral gene expression and replication. Recent reports have shown that RNA interference can be induced in mammalian cells by short interfering RNA duplexes (siRNA). Here we studied the effects of an HBV-specific 21-bp siRNA targeted to the surface antigen region (HBsAg), where three major viral mRNAs overlap, on HBV gene expression and replication both in a cell culture system and in a mouse model for HBV replication. Transfection of siRNA into HepG2.2.15 cells, which constitutively produce HBV particles, caused a significant reduction in viral RNA production that was accompanied by a >80% drop in the secretion of viral HBsAg and HBeAg into the medium. The effect of RNAi was tested in vivo in a mouse model that we have developed for HBV infection, which entails hydrodynamic injection of a plasmid bearing the HBV genome into tail veins of mice. Injection of the HBV plasmid induces viral replication and generation of HBV viral particles detectable in the mouse sera. Co-injection of the HBV plasmid together with siRNA caused a significant inhibition in the level of viral transcripts, viral antigens, and viral DNA detected in the livers and sera of the treated mice relative to control animals. Results suggest that siRNA is capable of inhibiting HBV replication in vivo and thus may constitute a new therapeutic strategy for HBV infection.


Subject(s)
Hepatitis B virus/genetics , Hepatitis B virus/physiology , RNA, Small Interfering/metabolism , Virus Replication , Animals , Blotting, Northern , Blotting, Southern , Cell Line , Disease Models, Animal , Dose-Response Relationship, Drug , Female , Humans , Immunohistochemistry , Kinetics , Liver/metabolism , Mice , Mice, Inbred BALB C , Plasmids/metabolism , Polymerase Chain Reaction , RNA Interference , RNA, Messenger/metabolism , Time Factors , Transfection
8.
Curr Opin Mol Ther ; 4(2): 102-9, 2002 Apr.
Article in English | MEDLINE | ID: mdl-12044030

ABSTRACT

A Trimera mouse is constructed through a three-step process. Firstly, a normal mouse host is rendered immuno-incompetent by a lethal split-dose total body irradiation. Secondly, the myeloid and erythroid lineages are reconstituted by transplantation of bone marrow cells from a genetically immune-deficient mouse donor. Thirdly. the resulting preconditioned mouse is transplanted with human cells or tissues that can be maintained in the foreign, yet supporting, environment for a considerable period of time. Immunization of Trimera mice, engrafted with human immune cells, induces a strong human immune response, thereby enabling generation of human therapeutic monoclonal antibodies (mAbs) via hybridoma technology. Transplantation of infected human tissue into the preconditioned mice results in the creation of Trimera mouse models for human diseases.


Subject(s)
Antibodies, Monoclonal/biosynthesis , Disease Models, Animal , Mice, Mutant Strains , Animals , Hepatitis B/immunology , Hepatitis B/pathology , Hepatitis B virus/immunology , Hepatitis C/immunology , Hepatitis C/pathology , Mice , Neoplasms/immunology , Neoplasms/pathology , Rats
9.
J Infect Dis ; 185(2): 153-61, 2002 Jan 15.
Article in English | MEDLINE | ID: mdl-11807688

ABSTRACT

The lack of small-animal models that are suitable for evaluation of agents used to treat infection with hepatitis C virus (HCV) severely hinders the assessment of potential new therapies for the disease. This study created such a model, termed the "HCV-Trimera" model. The HCV-Trimera model was developed by using lethally irradiated mice, reconstituted with SCID mouse bone marrow cells, in which human liver fragments infected ex vivo with HCV had been transplanted. Viremia (positive-strand HCV RNA levels) in HCV-Trimera mice peaked at approximately day 18 after liver transplantation, and an infection rate of 85% was reached. Viral replication in liver grafts was evidenced by the presence of specific negative-strand HCV RNA. The usefulness of this model for evaluation of anti-HCV agents was demonstrated by the ability of a small molecule (an HCV internal ribosomal entry site inhibitor) and an anti-HCV human monoclonal antibody (HCV AB(XTL)68) to reduce virus loads in HCV-Trimera mice in a dose-dependent manner.


Subject(s)
Antiviral Agents/therapeutic use , Disease Models, Animal , Hepatitis C/drug therapy , Animals , Hepatitis C/etiology , Hepatitis C/virology , Humans , Liver/virology , Mice , RNA, Viral/blood , Virus Replication
SELECTION OF CITATIONS
SEARCH DETAIL
...