Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 14 de 14
Filter
Add more filters










Publication year range
1.
Methods Mol Biol ; 635: 133-45, 2010.
Article in English | MEDLINE | ID: mdl-20552345

ABSTRACT

The utilization of macromolecules in therapy of cancer and other diseases is becoming increasingly relevant. Recent advances in molecular biology and biotechnology have made it possible to improve targeting and design of cytotoxic agents, DNA complexes, and other macromolecules for clinical applications. To achieve the expected biological effect of these macromolecules, in many cases, internalization to the cell cytosol is crucial. At an intracellular level, the most fundamental obstruction for cytosolic release of the therapeutic molecule is the membrane-barrier of the endocytic vesicles. Photochemical internalization (PCI) is a novel technology for release of endocytosed macromolecules into the cytosol. The technology is based on the use of photosensitizers located in endocytic vesicles that upon activation by light induces a release of macromolecules from their compartmentalization in endocytic vesicles. PCI has been shown to potentiate the biological activity of a large variety of macromolecules and other molecules that do not readily penetrate the plasma membrane, including type I ribosome-inactivating proteins (RIPs), gene-encoding plasmids, adenovirus, oligonucleotides, and the chemotherapeutic bleomycin. PCI has also been shown to enhance the treatment effect of targeted therapeutic macromolecules. The present protocol describes PCI of an epidermal growth factor receptor (EGFR)-targeted protein toxin (Cetuximab-saporin) linked via streptavidin-biotin for screening of targeted toxins as well as PCI of nonviral polyplex-based gene therapy. Although describing in detail PCI of targeted protein toxins and DNA polyplexes, the methodology presented in these protocols are also applicable for PCI of other gene therapy vectors (e.g., viral vectors), peptide nucleic acids (PNA), small interfering RNA (siRNA), polymers, nanoparticles, and some chemotherapeutic agents.


Subject(s)
Drug Delivery Systems/methods , Endocytosis/drug effects , Endocytosis/radiation effects , Photochemical Processes , Photosensitizing Agents/pharmacology , Antibodies, Monoclonal/chemistry , Antibodies, Monoclonal/metabolism , Antibodies, Monoclonal, Humanized , Biotinylation , Cell Line , Cetuximab , Cytosol/drug effects , Cytosol/metabolism , Cytosol/radiation effects , ErbB Receptors/metabolism , Light , Polyethyleneimine/chemistry , Polylysine/chemistry , Ribosome Inactivating Proteins, Type 1/metabolism , Saporins
2.
Methods Mol Biol ; 434: 171-81, 2008.
Article in English | MEDLINE | ID: mdl-18470645

ABSTRACT

Photochemical internalization (PCI) is a physico-chemical targeting method that enables light directed delivery of nucleic acids into cells. The technology is based on photosensitizers that localize in the membranes of endocytic vesicles. A light activation of the photosensitizers induces photochemical reactions that lead to rupture of the vesicular membranes. This results in the release of endocytosed compounds (e.g., nucleic acids) into the cell cytosol. Physico-chemical and biological targeting techniques can be combined to promote efficient and specific gene delivery to target cells. The present protocol describes PCI of epidermal growth factor receptor (EGFR)-targeted DNA polyplexes. The DNA polyplexes made are small (50-100 nm in diameter), and they contain polyethylenimine (PEI) conjugated with the EGF protein as a cell-binding ligand for EGFR-mediated endocytosis and polyethylene glycol (PEG) for masking the polyplex surface charge. PCI of such targeted PEG-PEI/DNA polyplexes enables high and EGFR-specific gene transfer activity in cells. Although describing in detail PCI of DNA polyplexes, the methodology presented in this protocol is also applicable for PCI of other gene therapy vectors (e.g. viral vectors), peptide nucleic acids (PNA), small interfering RNA (siRNA), and for vectors targeted to alternate cell surface receptors. Generally, PCI can be applied whenever 100% survival of the treated cell population is not required.


Subject(s)
DNA/administration & dosage , ErbB Receptors/metabolism , Gene Transfer Techniques , Photosensitizing Agents/chemistry , Polyethylene Glycols/chemistry , Polyethyleneimine/chemistry , DNA/chemistry , DNA/metabolism , Epidermal Growth Factor/metabolism , Genetic Vectors , Light , Transfection
3.
Curr Pharm Biotechnol ; 8(6): 362-72, 2007 Dec.
Article in English | MEDLINE | ID: mdl-18289045

ABSTRACT

The utilisation of macromolecules in the therapy of cancer and other diseases is becoming increasingly important. Recent advances in molecular biology and biotechnology have made it possible to improve targeting and design of cytotoxic agents, DNA complexes and other macromolecules for clinical applications. In many cases the targets of macromolecular therapeutics are intracellular. However, degradation of macromolecules in endocytic vesicles after uptake by endocytosis is a major intracellular barrier for the therapeutic application of macromolecules having intracellular targets of action. Photochemical internalisation (PCI) is a novel technology for the release of endocytosed macromolecules into the cytosol. The technology is based on the activation by light of photosensitizers located in endocytic vesicles to induce the release of macromolecules from the endocytic vesicles. Thereby, endocytosed molecules can be released to reach their target of action before being degraded in lysosomes. PCI has been shown to stimulate intracellular delivery of a large variety of macromolecules and other molecules that do not readily penetrate the plasma membrane, including type I ribosome-inactivating proteins (RIPs), DNA delivered as gene-encoding plasmids or by means of adenovirus or adeno-associated virus, peptide nucleic acids (PNAs) and chemotherapeutic agents such as bleomycin and in some cases doxorubicin. PCI of PNA may be of particular importance due to the low therapeutic efficacy of PNA in the absence of an efficient delivery technology and the 10-100-fold increased efficacy in combination with PCI. The efficacy and specificity of PCI of macromolecular therapeutics has been improved by combining the macromolecules with targeting moieties, such as the epidermal growth factor. In general, PCI can induce efficient light-directed delivery of macromolecules into the cytosol, indicating that it may have a variety of useful applications for site-specific drug delivery as for example in gene therapy, vaccination and cancer treatment.


Subject(s)
Drug Delivery Systems/methods , Macromolecular Substances , Pharmaceutical Preparations , Photosensitizing Agents , Animals , Endocytosis , Genetic Therapy/methods , Humans , Light , Macromolecular Substances/administration & dosage , Macromolecular Substances/chemistry , Molecular Structure , Pharmaceutical Preparations/administration & dosage , Pharmaceutical Preparations/chemistry , Photochemistry , Photosensitizing Agents/administration & dosage , Photosensitizing Agents/chemistry , Transport Vesicles/metabolism
4.
Cancer Biol Ther ; 5(11): 1511-20, 2006 Nov.
Article in English | MEDLINE | ID: mdl-17012835

ABSTRACT

Tumor targeting is an important issue in cancer gene therapy. We have developed a light-specific transduction method, named photochemical internalization (PCI), to enhance gene expression from adenoviral vectors selectively in illuminated areas. Tumor necrosis factor related apoptosis inducing ligand (TRAIL) has been shown to induce apoptosis in cancer cells, and the aim of this study was to investigate the potential of PCI to enhance transgene expression from AdhCMV-TRAIL and evaluate its impact on apoptotic induction in the two human colorectal cancer cell lines HCT116 and WiDr. PCI-mediated delivery of AdhCMV-TRAIL enabled an increased expression of TRAIL, induced a synergistic reduction in cell viability compared to the individual action of AdhCMV-TRAIL and photochemical treatment, and enhanced the induction of apoptosis demonstrated by an increase in cytoplasmic histone-associated DNA fragments, caspase-8 and caspase-3 activation, PARP cleavage and a decrease in the mitochondrial membrane potential. The synergistic effect could be related to the enhanced TRAIL expression in PCI-treated samples and a modest sensitization of the cancer cells to TRAIL induced apoptosis due to the photochemical treatment. Furthermore, an increased cleavage of Bid and a cell line dependent reduction in the expression levels of anti-apoptotic Bcl-2 family members were observed and could possibly contribute to the enhanced apoptotic level in samples exposed to the combined treatment. The presented results indicate that photochemically mediated delivery of AdhCMV-TRAIL allows a selective enhancement in cell killing, and suggest that PCI may be relevant and advantageous for therapeutic gene delivery in vivo.


Subject(s)
Colorectal Neoplasms/pathology , TNF-Related Apoptosis-Inducing Ligand/physiology , TNF-Related Apoptosis-Inducing Ligand/therapeutic use , Adenoviridae/genetics , Apoptosis , Cell Line, Tumor , Cell Survival , Cytomegalovirus/genetics , Genetic Vectors , Humans , Mitotic Index , Photochemistry , TNF-Related Apoptosis-Inducing Ligand/genetics
5.
J Pharmacol Exp Ther ; 319(2): 604-12, 2006 Nov.
Article in English | MEDLINE | ID: mdl-16902053

ABSTRACT

Drug resistance is a major problem for chemotherapy. Entrapment of anticancer drugs in endolysosomal compartments or active extrusions by plasma membrane proteins of the ATP-binding cassette (ABC) superfamily are important resistance mechanisms. This study evaluated photochemical internalization (PCI) of membrane-impermeable macromolecules that are not the target of ABC drug pumps for treating multidrug-resistant (MDR) cancer cells. We used the drug-sensitive uterine fibrosarcoma cell line MES-SA and its MDR, P-glycoprotein (P-gp)-overexpressing derivative MES-SA/Dx5 with the photosensitizer disulfonated meso-tetraphenylporphine (TPPS(2a)) and broad spectrum illumination. The PCI of doxorubicin, the ribosome-inactivating protein gelonin and adenoviral transduction were assessed in both cell lines, together with the uptake and excretion of TPPS(2a) and of two fluid phase markers easily detectable by fluorescence [lucifer yellow (LY) and fluorescein isothiocyanate (FITC)-dextran], as a model of gelonin uptake. Both cell lines were resistant to PCI of doxorubicin, but equally sensitive to PCI of gelonin, even though the endocytosis rates of LY and FITC-dextran were significantly lower in the MDR cells. In control studies, MES-SA/Dx5 cells were more resistant to photodynamic therapy (TPPS(2a) + light only). This was not mediated by P-gp, as there were no differences in the uptake and efflux of TPPS(2a) between the cell lines. After adenoviral infection, PCI enhanced gene delivery in both cell lines. In conclusion, PCI of macromolecular therapeutic agents that are not targets of P-gp is a novel therapeutic strategy to kill MDR cancer cells.


Subject(s)
Neoplasms/drug therapy , Photochemotherapy/methods , Photosensitizing Agents/therapeutic use , Porphyrins/therapeutic use , Adenoviridae/metabolism , Cell Line, Tumor , Dextrans/metabolism , Doxorubicin/pharmacology , Drug Resistance, Multiple , Drug Resistance, Neoplasm , Fluorescein-5-isothiocyanate/analogs & derivatives , Fluorescein-5-isothiocyanate/metabolism , Humans
6.
Photochem Photobiol Sci ; 5(4): 411-21, 2006 Apr.
Article in English | MEDLINE | ID: mdl-16583022

ABSTRACT

Photochemical internalization (PCI) enhances adenovirus (Ad) transgene expression in a variety of cell lines in vitro. However, measurements of the photochemical effect on transduction in multicellular environments are lacking. In this study, spheroids of DU 145 prostate cancer cells were used as a model to evaluate Ad serotype 5 (Ad5) transduction in a multicellular environment in response to PCI treatment. Furthermore, the Ad5 was coated with poly(2-methyl-acrylic acid 2-[(2-(dimethylamino)-ethyl)-methyl-amino]-ethyl ester) (pDAMA) to evaluate whether physicochemical properties such as charge and size of viral vectors affect transduction of photochemically treated spheroids. Spheroids incubated with photosensitizer TPPS(2a) (1 microg ml(-1)) and infected with adenovirus contained 3-fold higher percentage of reporter gene expressing cells after exposure to blue light (0.42 J cm(-2)) compared to no light, as analysed by flow cytometry of dissociated spheroids two days after treatment. The cells within the infected spheroids were further divided into three sections corresponding to the interior, intermediate and peripheral layers of the spheroids. This was performed by staining the spheroids with a diffusion-limited dye prior to dissociation. Transduction of cells within photochemically treated and untreated spheroids was heterogeneous, with a radial reduction of transgene expression towards the inner section of the spheroid. The coating of Ad with pDAMA induced up to 2-fold decrease in transduction of cells in the interior section of spheroids compared to uncomplexed Ad, while transduction of the peripheral section remained unchanged. The decrease in transduction could be related to reduced diffusion due to the size of the Ad-pDAMA complexes.


Subject(s)
Adenoviridae/chemistry , Genetic Therapy/methods , Photosensitizing Agents/pharmacology , Polymethacrylic Acids/chemistry , Porphyrins/pharmacology , Transduction, Genetic/methods , Adenoviridae/genetics , Cell Line, Tumor , Humans , Light , Male , Photochemistry , Photosensitizing Agents/metabolism , Porphyrins/metabolism , Prostatic Neoplasms , Spheroids, Cellular/drug effects , Spheroids, Cellular/radiation effects , Transgenes
7.
J Gene Med ; 8(6): 707-18, 2006 Jun.
Article in English | MEDLINE | ID: mdl-16518880

ABSTRACT

BACKGROUND: In the present study the physical targeting technique photochemical internalization (PCI) has been used in combination with adenovirus. We have previously shown that PCI enhances transgene expression from AdhCMV-lacZ, and the aim of the present study was to further increase the understanding of photochemically mediated adenoviral transduction. METHODS: Two colorectal carcinoma cell lines, WiDr and HCT116, were pre-incubated with the photosensitizer TPPS(2a) or methylene blue derivates (MBD) followed by infection with adenovirus and light exposure. Transgene expression was measured by flow cytometry. Real-time polymerase chain reaction (PCR) and fluorescence in situ hybridization (FISH) were used to quantify the level of viral DNA in the nuclei. Real-time PCR was also used to measure the level of beta-galactosidase mRNA in samples infected with AdhCMV-lacZ. RESULTS: Exposing TPPS(2a)-treated cells to light enhanced the quantity of viral DNA in the nucleus, the mRNA level of the transgene and the transgene expression compared to non-illuminated cells. The increased transgene expression was independent of the promoter used, but dependent on the time of light exposure and the cellular localization of the photosensitizer. CONCLUSIONS: The enhanced transgene expression observed after photochemical treatment is most likely not a result of one event, but more an interplay between various mechanisms. An increased level of adenoviral DNA in the nucleus and a dependency of endosomal localization of the photosensitizer to obtain enhanced transgene expression suggested that endosomal rupture facilitated the transport of adenoviruses to the nucleus.


Subject(s)
Adenoviridae/drug effects , Adenoviridae/isolation & purification , Cell Nucleus/drug effects , Cell Nucleus/radiation effects , Endosomes/metabolism , Photosensitizing Agents/pharmacology , Adenoviridae/genetics , Cell Line, Tumor , Cell Nucleus/virology , DNA, Viral/genetics , Gene Expression/radiation effects , Genome, Viral/genetics , Green Fluorescent Proteins/genetics , HCT116 Cells , Humans , Photosensitizing Agents/metabolism , RNA, Messenger/genetics , RNA, Messenger/metabolism , Time Factors , Transduction, Genetic , Transgenes , Tumor Cells, Cultured , beta-Galactosidase/genetics
8.
J Environ Pathol Toxicol Oncol ; 25(1-2): 521-36, 2006.
Article in English | MEDLINE | ID: mdl-16566739

ABSTRACT

Photochemical internalization (PCI) is a new technology, where certain photosensitizing substances (photosensitizers) are used to improve the utilization of macromolecules for cancer therapy, in a site-specific manner. Degradation of macromolecules in endocytic vesicles after uptake by endocytosis is a major intracellular barrier for the therapeutic application of molecules having intracellular targets of action. PCI is based on the light activation of photosensitizers specifically located in the membrane of endocytic vesicles inducing the rupture of this membrane upon illumination. Thereby endocytosed molecules can be released to reach their target of action before being degraded in lysosomes. This has been shown to enhance the biological activity of a large variety of macromolecules and other molecules that do not readily penetrate the plasma membrane, including type I ribosome-inactivating proteins (RIPs), immunotoxins, gene-encoding plasmids, adenovirus, peptidenucleic acids, and the chemotherapeuticum bleomycin. In several cases up to a 100-fold increase in biological activity has been observed. This article reviews the background and present status of PCI.


Subject(s)
Drug Delivery Systems , Endocytosis , Photochemotherapy , Animals , Genetic Therapy/methods , Humans , Light , Pharmaceutical Preparations/administration & dosage , Proteins/administration & dosage
9.
J Gene Med ; 8(3): 286-97, 2006 Mar.
Article in English | MEDLINE | ID: mdl-16342300

ABSTRACT

BACKGROUND: The development of methods for specific delivery of genes into target tissues is an important issue for the further progress of gene therapy. Biological and physical targeting techniques may be combined to redirect gene therapy vectors to specific cells and enhance gene transfer. METHODS: The polymer poly(2-(dimethylamino)ethyl methacrylate) (pDMAEMA) was conjugated with avidin or poly(ethylene glycol) (PEG) and complexed with adenovirus serotype 5 (Ad5). Targeting of polymer-coated Ad5 to the epidermal growth factor receptor (EGFR) was accomplished by the binding of biotin-EGF to pDMAEMA-avidin. A photochemical treatment procedure using photosensitizer and light was applied to increase transduction with EGFR-targeted viral complexes. RESULTS: pDMAEMA-avidin efficiently enhanced transduction through unspecific viral uptake into cells, while pDMAEMA-PEG provided charge shielding of the complexes and increased the specificity to EGFR when biotin-EGF ligands were used. Transduction of PEG-containing, EGFR-targeted viral complexes was inhibited by 66% in coxsackie and adenovirus receptor (CAR)-deficient RD cells and by 47% in CAR-expressing DU 145 cells in receptor antibody experiments. The photochemical treatment had a substantial effect on transduction, enhancing the percentage of reporter gene positive cells from 20% to 75% of the total viable RD cell population and from 10% to 70% in DU 145 cells. CONCLUSION: Photochemical treatment of cells infected with targeted viral vectors exhibiting a neutral surface charge is a potent method for enhancing transgene expression.


Subject(s)
ErbB Receptors/genetics , Gene Transfer Techniques , Genetic Therapy/methods , Adenoviridae/genetics , Avidin , Drug Carriers , ErbB Receptors/biosynthesis , Female , Gene Expression Profiling , Humans , Male , Methacrylates , Nylons , Photochemistry , Polyethylene Glycols , Prostatic Neoplasms/pathology , Rhabdomyosarcoma, Embryonal/pathology , Sarcoma/pathology , Transduction, Genetic , Tumor Cells, Cultured , Uterine Neoplasms/pathology
10.
Anticancer Res ; 25(1A): 291-7, 2005.
Article in English | MEDLINE | ID: mdl-15816550

ABSTRACT

BACKGROUND: Photodynamic therapy (PDT) and gene therapy protocols are separately under clinical evaluation for treatment of brain malignancies. Here, the potential of a novel combination technique, photo-induced delivery of macromolecules and genes to glioblastoma cells, is evaluated. MATERIALS AND METHODS: The photochemical effect on survival of GaMg and U-87Mg cells after incubation with the protein toxin gelonin, on transfection with a plasmid complexed to poly-L-lysine (PLL), and on transduction with adenovirus serotype 5 (Ad5) and adeno-associated virus type 5 (AAV5) vectors, were studied. RESULTS: Cytotoxicity of gelonin and gene transfer from plasmid/PLL complexes were considerably improved by photochemical treatment in both cell lines, while the light-inducible effect on Ad5 transduction was most pronounced in U-87Mg. For the first time, photochemical enhancement of AAV transduction is shown. A 4-fold increase in percentage positive cells was detected after photochemical treatment of AAV5-infected GaMg cells. However, in contrast to Ad5, AAV5 transduction of U-87Mg remained unaffected by light treatment, independently of viral dose, light dose and timing of the light treatment relative to the transduction period. CONCLUSION: Photochemical treatment is a versatile tool for macromolecular delivery to glioblastoma cells, however, the photochemical effect on gene transfer by viral vectors is highly dependent on the cell line and vector applied.


Subject(s)
Brain Neoplasms/therapy , Genetic Therapy/methods , Glioblastoma/therapy , Photochemotherapy/methods , Plant Proteins/administration & dosage , Adenoviridae/genetics , Adult , Brain Neoplasms/drug therapy , Brain Neoplasms/genetics , Brain Neoplasms/metabolism , Combined Modality Therapy , Female , Genetic Vectors/genetics , Glioblastoma/drug therapy , Glioblastoma/genetics , Glioblastoma/metabolism , Green Fluorescent Proteins/biosynthesis , Green Fluorescent Proteins/genetics , Humans , Plant Proteins/genetics , Polylysine/genetics , Ribosome Inactivating Proteins, Type 1 , Transduction, Genetic , Transfection
11.
Cancer Gene Ther ; 12(5): 439-48, 2005 May.
Article in English | MEDLINE | ID: mdl-15678152

ABSTRACT

The development of methods for efficient and specific delivery of therapeutic genes into target tissues is an important issue for further development of in vivo gene therapy. In the present study, the physical targeting technique, photochemical internalization (PCI), has been used together with adenovirus. The combination of PCI and adenoviral transduction has previously been shown to be favorable compared to adenovirus used alone, and the aim of this study was to verify the role of the adenoviral receptors and identify the uptake pathway used by adenoviral particles in photochemically treated cells. All examined cell lines showed augmented transduction efficiency after PCI-treatment, with a maximum of 13-fold increase in transgene expression compared to conventionally infected cells. Blocking of CAR induced a complete inhibition of PCI-enhanced transgene expression. However, photochemical treatment managed to enhance the transduction efficiency of the retargeted virus AdRGD-GFP showing also that the virus-CAR interaction is not vital for obtaining a photochemical effect on adenoviral transduction. Blocking the alpha(V)-integrins reduced the gene expression significantly in photochemically treated cells. Subjecting HeLa cells expressing negative mutant-dynamin to light treatment after infection gave no significant increase in gene transfer, while the gene transfer were enhanced seven-fold in cells with wild-type dynamin. Furthermore, chlorpromazine inhibited photochemical transduction in a dose-dependent manner, whereas Filipin III had no effect on the gene transfer. In summary, the data presented imply that adenoviral receptor binding is important and clathrin-mediated endocytosis is the predominant uptake mechanism for adenoviral particles in photochemically treated cells.


Subject(s)
Adenoviridae/genetics , Endocytosis , Genetic Vectors/genetics , Photochemistry/methods , Transduction, Genetic , Cell Line, Tumor , Clathrin/physiology , Coxsackie and Adenovirus Receptor-Like Membrane Protein , Humans , Integrin alphaV/physiology , Receptors, Virus/physiology
12.
Expert Opin Biol Ther ; 4(9): 1403-12, 2004 Sep.
Article in English | MEDLINE | ID: mdl-15335308

ABSTRACT

This article reviews a novel technology, named photochemical internalisation (PCI), for light-directed delivery of transgenes. Most gene therapy vectors are taken into the cell by endocytosis and, hence, are located in the endocytic vesicles. Although viral vectors have developed the means to escape from these vesicles, poor endosomal release is one of the major obstacles for non-viral vectors. PCI is a technology that allows liberation of the entrapped vectors carrying a gene in response to illumination. The method is based on chemical compounds (photosensitisers) that localise specifically in the membranes of endocytic vesicles and, following activation by light, induce the rupture of the vesicular membranes. The released transgenes can further be transferred to the nucleus, transcribed and translated. As gene liberation depends on light, enhancement of gene expression is achieved only at illuminated regions. PCI substantially improves gene transfer in vitro not only with non-viral gene vectors, but, surprisingly, also with adenoviruses and adeno-associated viruses. This article will review the background for the PCI technology and its role for gene delivery using both non-viral and viral vectors. Some aspects of the potential of PCI for site-specific gene delivery in therapeutic situations will also be discussed.


Subject(s)
Endocytosis/radiation effects , Endosomes/radiation effects , Gene Transfer Techniques , Genetic Vectors/administration & dosage , Intracellular Membranes/radiation effects , Photosensitizing Agents/pharmacology , Transgenes , Adenoviridae/genetics , Animals , Dependovirus/genetics , Gene Expression/radiation effects , Genes, Transgenic, Suicide , Genetic Vectors/genetics , Humans , Indoles/administration & dosage , Indoles/pharmacology , Intracellular Membranes/chemistry , Mice , Oligonucleotides, Antisense/administration & dosage , Organometallic Compounds/administration & dosage , Organometallic Compounds/pharmacology , Peptide Nucleic Acids/administration & dosage , Photochemistry , Photosensitizing Agents/administration & dosage , Porphyrins/administration & dosage , Porphyrins/pharmacology , Singlet Oxygen/pharmacology , Tumor Cells, Cultured , Xenograft Model Antitumor Assays
13.
Adv Drug Deliv Rev ; 56(1): 95-115, 2004 Jan 13.
Article in English | MEDLINE | ID: mdl-14706447

ABSTRACT

This article reviews a novel technology, named photochemical internalisation (PCI), for light-induced delivery of genes, proteins and many other classes of therapeutic molecules. Degradation of macromolecules in endocytic vesicles after uptake by endocytosis is a major intracellular barrier for the therapeutic application of macromolecules having intracellular targets of action. PCI is based upon the light activation of a drug (a photosensitizer) specifically locating in the membrane of endocytic vesicle inducing the rupture of this membrane upon illumination. Thereby endocytosed molecules can be released to reach their target of action before being degraded in lysosomes. The fact that this effect is induced by illumination means that the biological activity of the molecules can be activated at specific sites in the body, simply by illuminating the relevant region. We have used the PCI strategy to obtain light-induced delivery of a variety of molecules, including proteins, peptides, oligonucleotides, genes and low molecular weight drugs. In several cases, a >100-fold increase in biological activity has been observed.


Subject(s)
Drug Delivery Systems/methods , Genetic Therapy/methods , Neoplasms/drug therapy , Photochemotherapy , Photosensitizing Agents , Animals , Cell Line, Tumor , Humans , Light , Oligonucleotides/administration & dosage , Oligonucleotides/chemistry , Oligonucleotides/therapeutic use , Photochemotherapy/adverse effects , Photochemotherapy/methods , Photochemotherapy/trends , Photosensitizing Agents/administration & dosage , Photosensitizing Agents/chemistry , Photosensitizing Agents/therapeutic use
14.
Oftalmologia ; 56(1): 67-71, 2003.
Article in English | MEDLINE | ID: mdl-12886687

ABSTRACT

The utilisation of macromolecules in therapy of cancer and other diseases is becoming increasingly relevant. Recent advances in molecular biology and biotechnology have made it possible to improve targeting and design of cytotoxic agents or DNA complexes for clinical applications. To achieve the expected biological effect of these macromolecules in many cases internalization to the cell cytosol is crucial. A number of different methods for internalization of membrane impermeable molecules has been established, including electroporation, liposome fusion, antibodies/targeting ligands as protein carriers and the utilisation of various types of vectors such as cationic polymers and viruses, for gene therapy. Although new delivery systems have improved the cellular uptake of macromolecules, tissue penetration, cellular uptake and efficient transfer of the molecules into the cytosol of the target cell are still fundamental obstacles. At an intracellular level, the most fundamental obstruction for cytosolic release of the therapeutic molecule is the membrane-barrier of the endocytic vesicles. Photochemical internalization (PCI) is a novel technology for release of endocytosed macromolecules into the cytosol. The technology is based on the use of photosensitizers located in endocytic vesicles that upon activation by light induce a release of macromolecules from their compartmentalization in endocytic vesicles. PCI has been shown to potentiate the biological activity of a large variety of macromolecules and other molecules that do not readily penetrate the plasma membrane, including proteins, peptides, and DNA delivered as a complex with cationic polymers or incorporated in adenovirus. The basis as well as the utilization of this technology will be briefly reviewed in this paper.


Subject(s)
Photochemistry , Transport Vesicles/metabolism , Computer Graphics , Cytosol/metabolism , Drug Delivery Systems , Endosomes/metabolism , Humans , Macromolecular Substances , Neoplasms/drug therapy , Photosensitizing Agents/therapeutic use
SELECTION OF CITATIONS
SEARCH DETAIL
...