Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 27
Filter
Add more filters










Publication year range
1.
J Pharm Biomed Anal ; 49(4): 1014-20, 2009 May 01.
Article in English | MEDLINE | ID: mdl-19250790

ABSTRACT

The fully humanized Lewis-Y carbohydrate specific monoclonal antibody (mAb) IGN311 is currently tested in a passive immunotherapy approach in a clinical phase I trail and therefore regulatory requirements demand qualified assays for product analysis. To demonstrate the functionality of its Fc-region, the capacity of IGN311 to mediate complement dependent cytotoxicity (CDC) against human breast cancer cells was evaluated. The "classical" radioactive method using chromium-51 and a FACS-based assay were established and qualified according to ICH guidelines. Parameters evaluated were specificity, response function, bias, repeatability (intra-day precision), intermediate precision (operator-time different), and linearity (assay range). In the course of a fully nested design, a four-parameter logistic equation was identified as appropriate calibration model for both methods. For the radioactive assay, the bias ranged from -6.1% to -3.6%. The intermediate precision for future means of duplicate measurements revealed values from 12.5% to 15.9% and the total error (beta-expectation tolerance interval) of the method was found to be <40%. For the FACS-based assay, the bias ranged from -8.3% to 0.6% and the intermediate precision for future means of duplicate measurements revealed values from 4.2% to 8.0%. The total error of the method was found to be <25%. The presented data demonstrate that the FACS-based CDC is more accurate than the radioactive assay. Also, the elimination of radioactivity and the 'real-time' counting of apoptotic cells further justifies the implementation of this method which was subsequently applied for testing the influence of storage at 4 degrees C and 25 degrees C ('stability testing') on the potency of IGN311 drug product. The obtained results demonstrate that the qualified functional assay represents a stability indicating test method.


Subject(s)
Antibodies, Monoclonal/immunology , Antibodies/toxicity , Complement System Proteins/physiology , Lewis Blood Group Antigens/immunology , Algorithms , Biological Assay , Cell Line, Tumor , Cell Survival , Coloring Agents , Data Interpretation, Statistical , Female , Flow Cytometry , Humans , Linear Models , Reproducibility of Results , Risk Assessment
2.
Gene Ther ; 10(13): 1079-88, 2003 Jul.
Article in English | MEDLINE | ID: mdl-12808438

ABSTRACT

Systemic application of surface-shielded transferrin-polyethylenimine/DNA complexes leads to predominant DNA uptake and gene expression in Neuro2a tumors in syngeneic A/J mice. Similarly, high expression levels were found in Huh-7 and HepG2 human tumor xenografts in SCID mice after systemic application of surface-shielded EGF-PEG-PEI/DNA complexes. Significant DNA uptake but low gene expression were found in the M-3 melanoma while no DNA uptake and no gene expression were found in KB, 518A2, A549, and SW480 xenograft tumor models. To elucidate the reasons for these differences, the tumors were analyzed for vascularization and infiltration of macrophages. Neuro2a, Huh-7, and HepG2 tumors are well vascularized, with a high density of partially immature blood vessels and low numbers of infiltrating macrophages. The M-3 melanoma is well vascularized correlating with significant DNA uptake, however, necrosis and intensive infiltration by macrophages lead to rapid degradation of DNA. In contrast, the KB, 518A2, A549, and SW480 tumors are poorly vascularized, correlating with undetectable DNA uptake and gene expression. Using two different vector systems the data indicate that gene delivery to tumors in vivo is affected by tissue-dependent factors. Uptake of DNA into the tumor depends on vascularization of the tumor, while necrosis and macrophage infiltration may facilitate degradation of the DNA.


Subject(s)
Genetic Therapy/methods , Neoplasms/therapy , Transfection/methods , Transferrin/genetics , Animals , DNA/metabolism , Epidermal Growth Factor/genetics , Gene Expression , Humans , Injections, Intravenous , Macrophages/pathology , Mice , Mice, Inbred DBA , Mice, Inbred Strains , Mice, SCID , Neoplasms/blood supply , Neoplasms/pathology , Neoplasms, Experimental/blood supply , Neoplasms, Experimental/pathology , Neoplasms, Experimental/therapy , Neovascularization, Pathologic , Polyethyleneimine , Tissue Distribution , Tumor Cells, Cultured
3.
Gene Ther ; 10(9): 810-7, 2003 May.
Article in English | MEDLINE | ID: mdl-12704421

ABSTRACT

We investigated the efficacy and safety of the cationic polymer polyethylenimine (PEI) as a potential tool for intrauterine gene delivery into livers of fetal mice in the last trimester of pregnancy (E17.5). Using luciferase as a reporter gene, transferrin-conjugated and ligand-free PEI/DNA complexes (containing 3 microg DNA) with varying PEI-nitrogen/DNA-phosphate (N/P) ratios and different PEI forms, branched (800, 25 kDa) and linear (22 kDa), were compared with naked DNA. Transgene expression was measured 48 h after administration of PEI/DNA complexes or naked DNA. Highest luciferase activity (9.8 x 10(3) relative light units (RLU)/mg of tissue protein) was observed with ligand-free PEI22/DNA mixtures at N/P 6.0. In addition, this formulation was associated with very low toxicity as compared to the other PEI/DNA-injected groups. Using beta-galactosidase as a reporter gene, transfection of single, but also small, clusters of cells was demonstrated throughout the liver. Injection of 3 microg naked DNA resulted in an 11-fold lower transgene expression value (0.9 x 10(3) RLU/mg of tissue protein) as compared to PEI22/DNA complexes. However, the administration of higher concentrated naked DNA (9 microg) into fetal livers yielded expression levels of 3.2 x 10(4) RLU/mg of tissue protein, a more than three-fold increase compared to PEI22/DNA complexes. Furthermore, the gene transfer efficacy of concentrated naked DNA was approximately 40 times higher in fetuses than in adults (0.8 x 10(3) RLU/mg of tissue protein), indicating that fetal tissue is especially amenable to the uptake and expression of naked DNA.


Subject(s)
DNA/administration & dosage , Fetal Diseases/therapy , Genetic Therapy/methods , Liver/embryology , Liver/metabolism , Transfection/methods , 3T3 Cells , Animals , Animals, Newborn , Female , Gene Expression , Luciferases/analysis , Luciferases/genetics , Mice , Mice, Inbred CBA , Microscopy, Electron , Polyethyleneimine , Tissue Distribution
4.
Gene Ther ; 9(11): 731-5, 2002 Jun.
Article in English | MEDLINE | ID: mdl-12032698

ABSTRACT

We have developed surface-shielded ligand-polycation based gene delivery systems which are able to target gene expression to distant tumors after systemic application. Tumor-specific targeting is achieved by (1) incorporation of cell-binding ligands; and (2) shielding of the complexes from non-specific interactions with blood components and non-target cells. Shielding of polycation/DNA complexes can be achieved by coating with either polyethylene glycol or by incorporating the ligand transferrin at high densities. Following systemic application, surface-shielded DNA complexes coding for a highly active, yet highly toxic cytokine, tumor necrosis factor-alpha (TNFalpha), localized gene expression to distant tumors, resulting in hemorrhagic tumor necrosis and inhibition of tumor growth. TNFalpha activity was confined to the tumor without systemic TNF-related toxicity. These results indicate that targeted gene delivery may be an attractive strategy to use highly potent molecules in cancer treatment.


Subject(s)
Gene Targeting/methods , Genetic Therapy/methods , Neoplasms, Experimental/therapy , Tumor Necrosis Factor-alpha/genetics , Animals , Gene Expression , Genetic Engineering , Humans , Mice , Mice, Inbred BALB C , Mice, Inbred Strains , Polyethyleneimine , Transferrin/genetics
5.
Adv Drug Deliv Rev ; 53(3): 341-58, 2001 Dec 31.
Article in English | MEDLINE | ID: mdl-11744176

ABSTRACT

The polycation polyethylenimine (PEI) has recently been widely employed for the design of DNA delivery vehicles. Gene delivery using PEI involves condensation of DNA into compact particles, uptake into the cells, release from the endosomal compartment into the cytoplasm, and uptake of the DNA into the nucleus. Particularly for in vivo gene delivery, optimal coordination and timing between DNA complexation for protection of the DNA from nucleases and the disassembly of the complexes is essential. For in vivo application, DNA complexes have to pass a variety of anatomical and physiological barriers, and an environment of biological fluids and extracellular matrix before reaching their targets. Furthermore, targeted gene delivery is seriously hampered by non-specific interactions with non-target cells. Strategies have been developed to protect transfection complexes from non-specific interactions and to increase target specificity and gene expression.


Subject(s)
Drug Delivery Systems , Gene Targeting/methods , Polyethyleneimine/chemistry , Animals , DNA/administration & dosage , DNA/chemistry , Humans , Polyethyleneimine/chemical synthesis
6.
J Gene Med ; 3(4): 362-72, 2001.
Article in English | MEDLINE | ID: mdl-11529666

ABSTRACT

BACKGROUND: Efficient gene transfer is a major challenge for non-viral gene therapy. Understanding how non-viral vectors initiate gene expression could lead to the development of new future vectors with enhanced efficacy. METHODS: Linear or branched polyethylenimine (PEI)/DNA complexes were generated in varying salt conditions and their transfection efficiencies were compared in vitro and in vivo using reporter genes, luciferase and green fluorescent protein, and rhodamine labeled DNA (pGeneGrip). RESULTS: The transfection efficiency of linear PEI22/DNA in vitro was generally greater than that of branched PEI/DNA when complexes were generated in salt containing buffer. However, PEI complexes generated under salt-free conditions generally had low transfection activity in vitro. In contrast, PEI22/DNA salt-free complexes were highly active in vivo. Branched PEI/DNA and salt containing PEI22/DNA complexes were generally 10-100-fold less active than the salt-free PEI22/DNA complexes. Salt-free PEI22/DNA complexes were small, but subsequently grew into aggregates when salt was added. In contrast, PEI25/DNA complexes remained small even after salt was added under the same conditions. Furthermore, PEI22/pGeneGrips complexes formed large aggregates associated with the cell membrane, cytoplasm and nucleus, while branched PEI complexes remained as small distinct particles associated with the cell membrane or in the cytoplasm. CONCLUSIONS: Branched and linear PEI/DNA complexes differ in their ability to transfect cells. The greater efficiency of linear PEI might be due to an inherent kinetic instability under salt conditions. Understanding how to employ this kinetic instability of linear PEI could help in designing future vectors with greater flexibility and transfection efficiency in vivo.


Subject(s)
Genetic Vectors , Polyethyleneimine/administration & dosage , Transfection , Animals , Cell Division , DNA/genetics , Drug Carriers , Genes, Reporter , Genetic Therapy/methods , Green Fluorescent Proteins , Luciferases/genetics , Luminescent Proteins/analysis , Luminescent Proteins/genetics , Melanoma, Experimental , Mice , Recombinant Proteins/analysis , Structure-Activity Relationship , Transfection/methods
7.
Bioconjug Chem ; 12(4): 529-37, 2001.
Article in English | MEDLINE | ID: mdl-11459457

ABSTRACT

With the aim of generating gene delivery systems for tumor targeting, we have synthesized a conjugate consisting of polyethylenimine (PEI) covalently modified with epidermal growth factor (EGF) peptides. Transfection efficiency of the conjugate was evaluated and compared to native PEI in three tumor cell lines: KB epidermoid carcinoma cells, CMT-93 rectum carcinoma cells, and Renca-EGFR renal carcinoma cells. Depending on the tumor cell line, incorporation of EGF resulted in an up to 300-fold increased transfection efficiency. This ligand-mediated enhancement and competition with free EGF strongly suggested uptake of the complexes through the EGF receptor-mediated endocytosis pathway. Shielded particles being crucial for systemic gene delivery, we studied the effect of covalent surface modification of EGF-PEI/DNA complexes with a poly(ethylene glycol) (PEG) derivative. An alternative way for the formation of PEGylated EGF-containing complexes was also evaluated where EGF was projected away from PEI/DNA core complexes through a PEG linker. Both strategies led to shielded particles still able to efficiently transfect tumor cells in a receptor-dependent fashion. These PEGylated EGF-containing complexes were 10- to 100-fold more efficient than PEGylated complexes without EGF.


Subject(s)
DNA/metabolism , Epidermal Growth Factor/metabolism , Gene Transfer Techniques , Polyethyleneimine/metabolism , Transfection , Animals , DNA/chemistry , Endocytosis/physiology , Epidermal Growth Factor/chemistry , ErbB Receptors/metabolism , Gene Expression , Humans , KB Cells , Kidney Neoplasms/genetics , Luciferases/genetics , Macromolecular Substances , Mice , Polyethylene Glycols/chemical synthesis , Polyethyleneimine/chemical synthesis , Rectal Neoplasms/genetics , Tumor Cells, Cultured
8.
J Control Release ; 72(1-3): 165-70, 2001 May 14.
Article in English | MEDLINE | ID: mdl-11389995

ABSTRACT

Incorporation of the receptor binding ligands transferrin (Tf) or epidermal growth factor (EGF) into DNA/polyethylenimine (PEI) complexes was found to enhance gene transfer into tumor cell lines in a receptor-dependent manner. In systemic applications, the surface charge of DNA complexes dominated the in vivo characteristics of gene transfer. Administration of surface-shielded Tf-polycation/DNA complexes into the tail vein of A/J mice resulted in preferential gene delivery into distantly growing subcutaneous Neuro2a tumors. In contrast, application of positively charged DNA/PEI complexes directed gene transfer primarily to the lung. Two alternatives of masking the surface charge of complexes were accomplished. In the first case, shielding was obtained by covalently coating of DNA/Tf-PEI complexes with polyethylene glycol (PEG). Alternatively, incorporation of sufficient Tf protein into the DNA complexes resulted in charge shielding even without PEGylation. In the latter case lower-molecular weight polycations (25 kDa PEI for Tf-PEI complexes, or 32 kDa polylysine for AVET complexes) were used.


Subject(s)
DNA/administration & dosage , Neoplasms/therapy , Transfection/methods , Animals , DNA/pharmacology , Humans , Ligands , Mice , Polymers , Surface Properties
9.
Cancer Lett ; 168(2): 145-54, 2001 Jul 26.
Article in English | MEDLINE | ID: mdl-11403919

ABSTRACT

Tumor antigen pulsed dendritic cells (DCs) can induce anti-tumor immunity. We studied strategies for the reliable generation of such a tumor vaccine by functional maturation of DCs via interaction of CD40 with its ligand (CD40L, CD154). Exposure of immature DCs to CD40L transgenic cells, soluble recombinant human CD40L molecules or lipopolysaccharide induced expression of the co-stimulatory molecules, CD80 and CD86, and supported an allogeneic mixed leukocyte reaction. In contrast, the release of IL-12, an important mediator of anti-tumor immunity, and antigen-specific expansion and IFNgamma secretion of lymphocytes, was strongly triggered only by DCs exposed to CD40L transgenic cells.


Subject(s)
CD40 Ligand/genetics , CD40 Ligand/immunology , Dendritic Cells/immunology , Fibroblasts/physiology , Keratinocytes/physiology , Antigens, CD/biosynthesis , B7-1 Antigen/biosynthesis , B7-2 Antigen , CD40 Ligand/pharmacology , Cancer Vaccines/genetics , Cancer Vaccines/immunology , Cell Communication/immunology , Dendritic Cells/cytology , Fibroblasts/cytology , Humans , Interferon-gamma/metabolism , Interleukin-12/metabolism , Keratinocytes/cytology , Lipopolysaccharides/immunology , Lymphocyte Activation/immunology , Lymphocyte Culture Test, Mixed , Membrane Glycoproteins/biosynthesis , Plasmids/genetics , Recombinant Proteins/pharmacology , Signal Transduction/physiology , T-Lymphocytes/immunology , T-Lymphocytes/metabolism , Transfection , Transgenes , Tumor Cells, Cultured
10.
Gene Ther ; 8(1): 28-40, 2001 Jan.
Article in English | MEDLINE | ID: mdl-11402299

ABSTRACT

Systemic application of positively charged polycation/DNA complexes has been shown to result in predominant gene expression in the lungs. Targeting gene expression to other sites, eg distant tumors, is hampered by nonspecific interactions largely due to the positive surface charge of transfection complexes. In the present study we show that the positive surface charge of PEI (25 kDa branched or 22 kDa linear)/DNA complexes can be efficiently shielded by covalently incorporating transferrin at sufficiently high densities in the complex, resulting in a dramatic decrease in nonspecific interactions, eg with erythrocytes, and decreased gene expression in the lung. Systemic application of transferrin-shielded PEI/DNA complexes into A/J mice bearing subcutaneously growing Neuro2a tumors via the tail vein resulted in preferential (100- to 500-fold higher) luciferase reporter gene expression in distant tumors as compared with the major organs including the lungs. Tumor targeting is also demonstrated by DNA uptake and beta-galactosidase gene expression in tumor cells. Assessing DNA distribution following systemic application significant amounts of DNA were found in the liver and tumor. However, in the liver, DNA was mainly taken up by Kupffer cells and degraded without significant transgene expression. In the tumor, DNA was associated mainly with tumor cells and frequently found near structures which resemble primitive blood vessels.


Subject(s)
Gene Transfer Techniques , Genetic Therapy/methods , Neuroblastoma/therapy , Polyethyleneimine/metabolism , Animals , DNA/pharmacokinetics , Erythrocyte Aggregation , Female , Fluorescent Antibody Technique , Gene Expression Regulation, Neoplastic , Mice , Mice, Inbred A , Molecular Weight , Neoplasm Transplantation , Neuroblastoma/metabolism , Polyethyleneimine/chemistry , Transferrin/genetics , Transferrin/metabolism , Transgenes/genetics , Tumor Cells, Cultured
11.
Cancer Lett ; 161(2): 241-50, 2000 Dec 20.
Article in English | MEDLINE | ID: mdl-11090975

ABSTRACT

We explored the potential of the xenogenization concept as an adjuvant procedure in anti-tumor immunity. To mediate effective loading we used polyarginine (pArg) molecules of various degrees of polymerization, cationic liposomes, or chimeric molecules of transferrin (Tf) and the polycation polyethyleneimine (PEI). Tetanus toxoid (TT) was loaded onto primary human leukemia cells, culture adapted primary human neuroblastoma cells, and human lymphoblastoid cell lines (LCLs) with high efficiency by all procedures. Trypsin treatment of loaded cells provided evidence that only liposomes and Tf-PEI mediated internalization of TT. Lymphocytes primed with xenogenized LCLs and challenged with unmodified LCLs showed increased IFNgamma secretion compared with lymphocytes primed with non-xenogenized LCLs.


Subject(s)
Leukemia, Lymphoid/drug therapy , Tetanus Toxoid/pharmacology , B-Lymphocytes/immunology , B-Lymphocytes/metabolism , Blotting, Western , Cations/pharmacology , Flow Cytometry , Fluorescein-5-isothiocyanate/pharmacology , Humans , Interferon-gamma/metabolism , Leukemia, Lymphoid/immunology , Liposomes/pharmacology , Lymphocyte Activation , Neuroblastoma/drug therapy , Neuroblastoma/immunology , Peptide Biosynthesis , Peptides/metabolism , Polyethyleneimine/pharmacology , Precipitin Tests , Transferrin/chemistry , Transferrin/pharmacology , Trypsin/pharmacology , Tumor Cells, Cultured
12.
Cancer Gene Ther ; 7(6): 870-8, 2000 Jun.
Article in English | MEDLINE | ID: mdl-10880017

ABSTRACT

Vaccination using well-characterized allogeneic tumor cell lines expressing standardized doses of immunostimulatory cytokines is an attractive alternative for autologous gene-transfected tumor cell vaccines. In the present study, we show that vaccination with irradiated allogeneic K1 735 (H-2k) or B16F10 (H-2b) melanoma cells induces a moderate degree of cross-protection against the M-3 melanoma (H-2d) in DBA/2 mice. Cross-protection against the syngeneic tumor was markedly improved when the allogeneic vaccines were transfected with the interleukin-2 (IL-2) gene. The IL-2 gene-modified allogeneic vaccines were effective for prophylactic vaccination against subsequent tumor challenge and for therapeutic vaccination against pre-existing tumor deposits, with efficacies that were comparable with that of the IL-2 gene-modified syngeneic vaccines. Cross-protection correlated with the cytotoxic activity of splenocytes against M-3 targets. Allogeneic vaccination was not effective in another model, against the B16F10 melanoma in C57BL/6 mice, irrespective of genetic modification with the IL-2 or granulocyte-macrophage colony-stimulating factor genes.


Subject(s)
Cancer Vaccines/administration & dosage , Interleukin-2/genetics , Melanoma, Experimental/prevention & control , Animals , Female , Follow-Up Studies , Injections, Subcutaneous , Interleukin-2/immunology , Melanoma, Experimental/immunology , Mice , Mice, Inbred C57BL , Mice, Inbred DBA , Spleen/cytology , Spleen/pathology , Survival Analysis , Transduction, Genetic , Transplantation, Homologous , Transplantation, Isogeneic , Tumor Cells, Cultured/radiation effects , Tumor Cells, Cultured/transplantation , Vaccination
13.
Pharm Res ; 17(1): 42-8, 2000 Jan.
Article in English | MEDLINE | ID: mdl-10714607

ABSTRACT

PURPOSE: Liposomal systems may be useful as a cytokine supplement in tumor cell vaccines by providing a cytokine reservoir at the antigen presentation site. Here, we examined the effect of liposome incorporation of mIFNgamma on its potency as adjuvant in an established tumor cell vaccination protocol in the murine B16 melanoma model. Adjuvanticity of the mIFNgamma-liposomes was compared to that achieved by mIFNgamma-gene transfection of the B16 tumor cells. Furthermore, we studied whether liposomal incorporation of mIFNgamma indeed increases the residence time of the cytokine at the vaccination site. METHODS: C57B1/6 mice were immunized with i) irradiated IFNgamma-gene transfected B16 melanoma cells or ii) irradiated wild type B16 cells supplemented with (liposomal) mIFNgamma, followed by a challenge with viable B16 cells. The residence time of the (liposomal) cytokine at the subcutaneous (s.c.) vaccination site was monitored using radiolabeled mIFNgamma and liposomes. RESULTS: Immunization with irradiated tumor cells admixed with liposomal mIFNgamma generated comparable protection against B16 challenge as immunization with mIFNgamma-gene modified tumor cells. Irradiated tumor cells admixed with soluble mIFNgamma did not generate any protective responses. Radiolabeling studies indicated that free mIFNgamma rapidly cleared from the s.c. injection site. Association of [125I]-mIFNgamma with liposomes increased the local residence time substantially: liposomal association of mIFNgamma resulted in a prolonged local residence time of the cytokine as reflected by a 4-fold increase of the area under the curve. The amount of released cytokine in the optimal dose range corresponds to the amount released by the gene-transfected cells. Moderate but significant CTL-activity against B16 cells was found for mice immunized with irradiated cells supplemented with mIFNgamma-liposomes compared to untreated control animals. CONCLUSIONS: Prolonged presence of mIFNgamma at the site of antigen presentation is crucial for the generation of systemic immune responses in the B16 melanoma model. These studies show that liposomal encapsulation of cytokines is an attractive strategy for paracrine cytokine delivery in tumor vaccine development.


Subject(s)
Adjuvants, Immunologic/administration & dosage , Cancer Vaccines/immunology , Interferon-gamma/administration & dosage , Animals , Cancer Vaccines/administration & dosage , Drug Carriers , Female , Interferon-gamma/genetics , Liposomes , Melanoma, Experimental/therapy , Mice , Mice, Inbred C57BL , T-Lymphocytes, Cytotoxic/immunology , Transfection , Vaccination
14.
Gene Ther ; 6(4): 595-605, 1999 Apr.
Article in English | MEDLINE | ID: mdl-10476219

ABSTRACT

We investigated the in vitro and in vivo properties of DNA/transferrin-polyethylenimine (800 kDa) complexes before and after covalent coupling of poly(ethylene glycol) (PEG). Upon incubation with plasma, the positively charged non-PEGylated DNA complexes form aggregates. Plasma proteins such as IgM, fibrinogen, fibronectin and complement C3 were found to bind to non-PEGylated DNA complexes. At DNA concentrations relevant for in vivo gene delivery a strong aggregation of erythrocytes was also observed. PEGylation of the complexes strongly reduces plasma protein binding and erythrocyte aggregation. Furthermore, PEGylated complex size was stabilized and had a reduced surface charge. Prolonged circulation in the blood of the PEGylated complexes was also observed when injected intravenously. In tumor bearing mice, application of non-PEGylated complexes through the tail vein resulted in reporter gene expression in tail and lung, but severe toxicity was observed in some mice. In contrast, PEGylated complexes mediated reporter gene transfer to the tumor without significant toxicity.


Subject(s)
Genetic Therapy/methods , Genetic Vectors/administration & dosage , Neoplasms, Experimental/therapy , Polyethylene Glycols/administration & dosage , Polyethyleneimine , Transfection/methods , Animals , Biophysical Phenomena , Biophysics , Blood Proteins/metabolism , Cell Line , DNA/metabolism , Erythrocytes/metabolism , Female , Gene Expression , Genetic Vectors/metabolism , Luciferases/genetics , Mice , Mice, Inbred Strains , Neoplasm Transplantation
15.
Int J Pharm ; 183(1): 33-6, 1999 Jun 10.
Article in English | MEDLINE | ID: mdl-10361150

ABSTRACT

Subcutaneous vaccination of C57bl/6 mice with irradiated B16 melanoma cells supplemented with liposomal interleukin-2 (IL2) or murine interferon-gamma (mIFNgamma), resulted in systemic protection in 50% of the animals, against a subsequent tumor cell challenge in a dose dependent manner. The protective efficacy was comparable to the efficacy of cytokine gene-modified cells as tumor vaccine, whereas irradiated B16 cells supplemented with soluble cytokine did not result in protective responses. In vivo evidence was obtained that the beneficial effects mediated by liposome incorporation of the cytokine are the result of a depot function of the liposomal cytokine supplement at the vaccination site. In can be concluded that liposomal delivery of cytokines offers an attractive alternative to cytokine-gene transfection of tumor cells for therapeutic vaccination protocols.


Subject(s)
Cancer Vaccines/immunology , Interferon-gamma/administration & dosage , Interleukin-2/administration & dosage , Liposomes/administration & dosage , Melanoma, Experimental/immunology , Animals , Interferon-gamma/genetics , Interleukin-2/genetics , Mice , Mice, Inbred C57BL , Transfection , Vaccination
16.
J Drug Target ; 7(4): 293-303, 1999 Dec.
Article in English | MEDLINE | ID: mdl-10682908

ABSTRACT

We describe the comparison of non-viral polycation transfection reagents, adenovirus-enhanced transferrinfection (AVET), polyethylenimine (PEI800) and transferrin-conjugated PEI800 (Tf-PEI800) in their ability to transfect murine and primary human melanoma cell lines. Expression of a reporter gene, cell surface marker and secreted protein (interleukin-2) was assessed for each vector system. Testing for luciferase reporter gene expression in murine and primary human cell lines, AVET and Tf-PEI800, both showed high levels of expression and comparable activity. Furthermore, when the melanoma cell line B16F10 was transfected with a cell surface marker up to approximately 97% of the cells expressed the protein on the cell surface. Assessing the levels of secreted IL-2 in murine cell lines, AVET/IL-2, Tf-PEI800/IL-2 and PEI800/IL-2 all expressed high levels of the cytokine (up to 20 microg IL-2/10(6) cells/24 h). In primary human melanoma cell lines, AVET/IL-2 transfected cells secreted more IL-2 than cells transfected with either Tf-PEI800/IL-2 or PEI800/IL-2. In murine melanoma cell culture experiments, positively charged PEI800/DNA and Tf-PEI800/DNA complexes gave similar transfection efficiencies. However, when subcutaneous tumors in mice were injected with the luciferase reporter gene complexed with either Tf-PEI800 or AVET, higher transfection activity was measured in the tumors as compared to ligand free PEI800/DNA complexes.


Subject(s)
Cations/chemistry , DNA/metabolism , Drug Delivery Systems/methods , Genetic Vectors/genetics , Melanoma/metabolism , Transferrin/genetics , Adenoviridae/chemistry , Animals , Cell Line , Cytokines/metabolism , Genes, Reporter/genetics , Humans , Interleukin-2/metabolism , Luciferases/genetics , Membrane Proteins/metabolism , Mice , Polyethyleneimine/chemistry , Transfection/genetics
17.
J Gene Med ; 1(2): 111-20, 1999.
Article in English | MEDLINE | ID: mdl-10738575

ABSTRACT

BACKGROUND: Efficient and target-specific in vivo gene delivery is a major challenge in gene therapy. Compared to cell culture application, in vivo gene delivery faces a variety of additional obstacles such as anatomical size constraints, interactions with biological fluids and extracellular matrix, and binding to a broad variety of non-target cell types. METHODS: Polycation-based vectors, including adenovirus-enhanced transferrinfection (AVET) and transferrin-polyethylenimine (Tf-PEI), were tested for gene delivery into subcutaneously growing tumors after local and systemic application. DNA biodistribution and reporter gene expression was measured in the major organs and in the tumor. RESULTS: Gene transfer after intratumoral application was 10-100 fold more efficient with Tf-PEI/DNA or AVET complexes in comparison to naked DNA. Targeted gene delivery into subcutaneously growing tumors after systemic application was achieved using electroneutral AVET complexes and sterically stabilized PEGylated Tf-PEI/DNA complexes, whereas application of positively charged polycation/DNA complexes resulted in predominant gene expression in the lungs and was associated by considerable toxicity. CONCLUSION: For systemic application, the physical and colloidal parameters of the transfection complexes, such as particle size, stability, and surface charge, determine DNA biodistribution, toxicity, and transfection efficacy. By controlling these parameters, DNA biodistribution and gene expression can be targeted to different organs.


Subject(s)
Cancer Vaccines/administration & dosage , Genetic Therapy , Vaccines, DNA/administration & dosage , Adenoviridae/genetics , Animals , Cancer Vaccines/chemistry , Drug Stability , Electrochemistry , Female , Gene Expression , Gene Targeting , Gene Transfer Techniques , Genes, Reporter , Genetic Vectors , Mice , Mice, Inbred A , Mice, Inbred DBA , Neoplasms, Experimental/genetics , Neoplasms, Experimental/therapy , Particle Size , Polyamines , Polyelectrolytes , Transfection , Vaccines, DNA/chemistry
18.
Clin Cancer Res ; 4(8): 1881-6, 1998 Aug.
Article in English | MEDLINE | ID: mdl-9717815

ABSTRACT

Vaccination with tumor cells genetically engineered to produce interleukin (IL)-2 is an attractive strategy to enhance antitumor immune responses. The improved antitumor immunity upon vaccination with IL-2 gene-modified tumor cells may be due to the prolonged presence of the cytokine at the vaccination site. Because liposomes have been used for sustained delivery of a variety of agents, we compared the protective effect of vaccines consisting of IL-2 gene-modified B16 melanoma cells to that of vaccines composed of IL-2 liposomes and irradiated melanoma cells. The results indicate that both approaches equally protect against a lethal challenge with B16 melanoma cells. More than 20% of the protected animals developed vitiligo at the vaccination and/or tumor challenge site.


Subject(s)
Cancer Vaccines/administration & dosage , Interleukin-2/administration & dosage , Melanoma, Experimental/genetics , Melanoma, Experimental/immunology , Transfection , Animals , Female , Immunity, Innate , Interleukin-2/genetics , Interleukin-2/immunology , Liposomes , Melanoma, Experimental/prevention & control , Mice , Mice, Inbred C57BL
19.
Cytokines Cell Mol Ther ; 4(2): 95-103, 1998 Jun.
Article in English | MEDLINE | ID: mdl-9681248

ABSTRACT

Murine melanoma cells were engineered to express interleukin-2 (IL-2), interferon-gamma (IFN-gamma) or both cytokines at various dose levels by means of the adenovirus-enhanced transferrinfection (AVET) method. The gene-modified cells were tested for their potency to induce an antitumor immune response in two experimental settings with different tumor load. In a prophylactic vaccination model, both IL-2 and IFN-gamma showed a dose-dependent protection against tumor cell challenge in two melanoma models. In the therapeutic vaccination model, where mice with measurable tumors were treated, immunization with IL-2 or IFN-gamma gene-modified cells led to complete tumor regression in 30% or 20% of the tumor-bearing animals respectively. The combination of IL-2 + IFN-gamma resulted in complete tumor regression in up to 50% of the tumor-bearing mice.


Subject(s)
Cancer Vaccines/administration & dosage , Interferon-gamma/genetics , Interleukin-2/genetics , Melanoma/prevention & control , Melanoma/therapy , Animals , Cancer Vaccines/genetics , Disease Models, Animal , Female , Genetic Therapy/methods , Melanoma/immunology , Mice , Mice, Inbred C57BL , Mice, Inbred DBA , T-Lymphocytes, Cytotoxic/immunology , Transfection , Tumor Cells, Cultured , Vaccination
20.
Gene Ther ; 5(1): 31-9, 1998 Jan.
Article in English | MEDLINE | ID: mdl-9536262

ABSTRACT

Cytokine-induced killer (CIK) cells have been shown to eradicate established tumors in a SCID mouse-human lymphoma model. CIK cells depend on exogenous addition of cytokines such as interleukin-2 (IL-2), interleukin-7 (IL-7) or interleukin-12 (IL-12) for proliferation. In this study, we used the adenovirus-enhanced CD3 receptor-mediated gene transfer for transfection with the IL-7 gene. An episomally replicating plasmid was used containing cDNA of the human IL-7 gene under the control of a CMV promoter for transfection of CIK cells. Biosynthesis of IL-7 was demonstrated by RT-PCR, an enzyme-linked immunosorbent assay (ELISA) and using a bioassay. Transfected cells produced IL-7 in the range between 200 and 1100 pg/10(6) cells in 24 h. IL-7 was shown to be biologically active, since transfected CIK cells showed an improved proliferation rate as compared with nontransfected cells. Expression of IL-7 altered the secretion of other cytokines by CIK cells, in particular the production of TNF alpha increased after transfection. In contrast, nontransfected CIK cells fed with IL-7 showed no increase in TNF alpha secretion. No significant differences were found in expression of surface antigens linked to the cytotoxic activity of CIK cells. Cytotoxic activity against various tumor cell lines (eg renal cell carcinoma, malignant melanoma and colon carcinoma) was tested. Transfected cells possessed a significantly higher cytotoxic activity as compared with nontransfected cells. Receptor-mediated gene transfer effectively delivers expression plasmids for therapeutic genes into CIK cells and CIK cells transfected with an IL-7 gene expression construct may be valuable for adoptive immunotherapy.


Subject(s)
Genetic Therapy/methods , Interleukin-7/genetics , Killer Cells, Natural/physiology , Neoplasms/therapy , Transfection/methods , Adenoviridae/genetics , Biological Assay , CD3 Complex/metabolism , CD56 Antigen/metabolism , Cell Division , Cytotoxicity Tests, Immunologic , Enzyme-Linked Immunosorbent Assay , Genetic Vectors , Humans , Interleukin-7/biosynthesis , Killer Cells, Natural/immunology , Polymerase Chain Reaction , Tumor Necrosis Factor-alpha/biosynthesis
SELECTION OF CITATIONS
SEARCH DETAIL