Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 12 de 12
Filter
Add more filters










Publication year range
1.
Oncotarget ; 7(15): 19997-20015, 2016 Apr 12.
Article in English | MEDLINE | ID: mdl-26918352

ABSTRACT

The knowledge on how tumor-associated stroma influences efficacy of anti-cancer therapy just started to emerge. Here we show that lung fibroblasts reduce melanoma sensitivity to the BRAF inhibitor (BRAFi) vemurafenib only if the two cell types are in close proximity. In the presence of fibroblasts, the adjacent melanoma cells acquire de-differentiated mesenchymal-like phenotype. Upon treatment with BRAFi, such melanoma cells maintain high levels of phospho ribosomal protein S6 (pS6), i.e. active mTOR signaling, which is suppressed in the BRAFi sensitive cells without stromal contacts. Inhibitors of PI3K/mTOR in combination with BRAFi eradicate pS6high cell subpopulations and potentiate anti-cancer effects in melanoma protected by the fibroblasts. mTOR and BRAF co-inhibition also delayed the development of early-stage lung metastases in vivo. In conclusion, we demonstrate that upon influence from fibroblasts, melanoma cells undergo a phenotype switch to the mesenchymal state, which can support PI3K/mTOR signaling. The lost sensitivity to BRAFi in such cells can be overcome by co-targeting PI3K/mTOR. This knowledge could be explored for designing BRAFi combination therapies aiming to eliminate both stroma-protected and non-protected counterparts of metastases.


Subject(s)
Fibroblasts/pathology , Melanoma/pathology , Mesoderm/pathology , Phosphatidylinositol 3-Kinases/metabolism , Protein Kinase Inhibitors/pharmacology , Proto-Oncogene Proteins B-raf/antagonists & inhibitors , TOR Serine-Threonine Kinases/metabolism , Animals , Apoptosis , Cell Proliferation , Fibroblasts/drug effects , Fibroblasts/metabolism , Humans , Melanoma/drug therapy , Melanoma/metabolism , Mesoderm/drug effects , Mesoderm/metabolism , Mice, Nude , Signal Transduction/drug effects , Stromal Cells/drug effects , Stromal Cells/metabolism , Stromal Cells/pathology , Tumor Cells, Cultured , Xenograft Model Antitumor Assays
2.
Cancer Biol Ther ; 12(1): 47-58, 2011 Jul 01.
Article in English | MEDLINE | ID: mdl-21508672

ABSTRACT

Melanoma is a highly aggressive malignant tumor with an exceptional ability to develop resistance and no curative therapy is available for patients with distant metastatic disease. The inhibitor of apoptosis protein (IAP) family has been related to therapy resistance in cancer. We examined the importance of the IAPs in the resistance to the commonly used chemotherapeutic agent dacarbazine (DTIC) and the apoptosis inducer TRAIL (TNF-related apoptosis inducing ligand) in malignant melanoma. The data presented show that the expression of IAPs is universal, concomitant and generally high in melanoma cell lines and in patient samples. Depleting IAP expression by siRNA tended to reduce cell viability, with XIAP reduction being the most efficient in all four cell lines examined (FEMX-1, LOX, SKMEL-28 and WM115). The combined treatment of XIAP siRNA and DTIC showed a weak improvement in two of four cell lines, while all four cell lines showed enhanced sensitivity towards TRAIL (AdhCMV-TRAIL) after XIAP depletion. In addition, cIAP-1, cIAP-2 and survivin down-regulation sensitized to TRAIL treatment in several of the cell lines. Cells exposed to TRAIL and XIAP siRNA showed increased DNA-fragmentation and cleavage of Bid, procaspase-8, -9, -7 and -3 and PARP, and change in the balance between pro- and anti-apoptotic proteins, indicating an enhanced level of apoptosis. Furthermore, the combined treatment reduced the ability of melanoma cells to engraft and form tumors in mice, actualizing the combination for future therapy of malignant melanoma.


Subject(s)
Antineoplastic Agents, Alkylating/pharmacology , Dacarbazine/pharmacology , Inhibitor of Apoptosis Proteins/metabolism , Melanoma/drug therapy , TNF-Related Apoptosis-Inducing Ligand/metabolism , Apoptosis , BH3 Interacting Domain Death Agonist Protein/metabolism , Baculoviral IAP Repeat-Containing 3 Protein , Caspases/metabolism , Cell Line, Tumor , Cell Survival/drug effects , DNA Fragmentation , Drug Resistance, Neoplasm , Humans , Inhibitor of Apoptosis Proteins/drug effects , Inhibitor of Apoptosis Proteins/genetics , Melanoma/metabolism , Melanoma/pathology , Poly(ADP-ribose) Polymerases/metabolism , RNA, Small Interfering , Survivin , TNF-Related Apoptosis-Inducing Ligand/genetics , Tumor Stem Cell Assay , Ubiquitin-Protein Ligases , X-Linked Inhibitor of Apoptosis Protein/drug effects , X-Linked Inhibitor of Apoptosis Protein/genetics
3.
PLoS One ; 5(5): e10731, 2010 May 20.
Article in English | MEDLINE | ID: mdl-20505780

ABSTRACT

BACKGROUND: Malignant melanoma is an exceptionally aggressive, drug-resistant and heterogeneous cancer. Recently it has been shown that melanoma cells with high clonogenic and tumourigenic abilities are common, but markers distinguishing such cells from cells lacking these abilities have not been identified. There is therefore no definite evidence that an exclusive cell subpopulation, i.e. cancer stem cells (CSC), exists in malignant melanoma. Rather, it is suggested that multiple cell populations are implicated in initiation and progression of the disease, making it of importance to identify subpopulations with elevated aggressive properties. METHODS AND FINDINGS: In several other cancer forms, Aldehyde Dehydrogenase (ALDH), which plays a role in stem cell biology and resistance, is a valuable functional marker for identification of cells that show enhanced aggressiveness and drug-resistance. Furthermore, the presence of ALDH(+) cells is linked to poor clinical prognosis in these cancers. By analyzing cell cultures, xenografts and patient biopsies, we showed that aggressive melanoma harboured a large, distinguishable ALDH(+) subpopulation. In vivo, ALDH(+) cells gave rise to ALDH(-) cells, while the opposite conversion was rare, indicating a higher abilities of ALDH(+) cells to reestablish tumour heterogeneity with respect to the ALDH phenotype. However, both ALDH(+) and ALDH(-) cells demonstrated similarly high abilities for clone formation in vitro and tumour initiation in vivo. Furthermore, both subpopulations showed similar sensitivity to the anti-melanoma drugs, dacarbazine and lexatumumab. CONCLUSIONS: These findings suggest that ALDH does not distinguish tumour-initiating and/or therapy-resistant cells, implying that the ALDH phenotype is not associated with more-aggressive subpopulations in malignant melanoma, and arguing against ALDH as a "universal" marker. Besides, it was shown that the ability to reestablish tumour heterogeneity is not necessarily linked to the more aggressive phenotype.


Subject(s)
Aldehyde Dehydrogenase/metabolism , Melanoma/enzymology , Melanoma/pathology , Animals , Biopsy , Cell Line, Tumor , Cell Proliferation , Cell Separation , Clone Cells , Humans , Melanoma/drug therapy , Mice , Phenotype , Receptors, TNF-Related Apoptosis-Inducing Ligand/metabolism , Xenograft Model Antitumor Assays
4.
J Gene Med ; 9(6): 440-51, 2007 Jun.
Article in English | MEDLINE | ID: mdl-17410615

ABSTRACT

BACKGROUND: Currently, dacarbazine (DTIC) is the only approved systemic treatment for metastatic malignant melanoma. However, the modest treatment effect encourages studies on novel therapeutic molecules, delivery systems and combination therapies. Full-length TRAIL, delivered from an adenoviral vector (Ad-hTRAIL), was studied in combination with DTIC or the histone deacetylase inhibitor suberoylanilide hydroxamic acid (SAHA) in human melanoma cell lines. METHODS: The cytotoxic potential of the combination treatments was assessed by cell viability measurements and CalcuSyn analysis. Involvement of apoptosis was analyzed by TUNEL staining, mitochondrial membrane potential measurements, and activation and expression levels of caspases and other mediators of apoptosis. RESULTS: Ad-hTRAIL in combination with DTIC or SAHA resulted in additive or synergistic growth inhibition compared to each treatment used as single agent. Both combinations augmented apoptosis, which was mediated through the death receptor (DR) pathway by enhanced activation of caspase-8, and through increased loss of mitochondrial integrity. Provoked cleavage of Bid, which bridges the extrinsic and intrinsic apoptosis pathways, and downregulation of the anti-apoptotic mediators Bcl-X(L), Mcl-1 and XIAP (but not Bcl-2) were critical contributing factors. Increased levels of DR4 and DR5 were not a common underlying mechanism as DTIC did not affect the levels of either of the receptors. However, SAHA-induced expression of DR4 may have reduced the TRAIL resistance in the SKMEL-28 cell line. CONCLUSION: Administration of Ad-hTRAIL in combination with DTIC or SAHA enhances apoptosis in human melanoma cell lines, and suggests that the therapeutic potential of such treatment strategies should be further evaluated for possible clinical use.


Subject(s)
Adenoviridae/genetics , Apoptosis/drug effects , Dacarbazine/pharmacology , Hydroxamic Acids/pharmacology , Melanoma/pathology , Mitochondria/drug effects , TNF-Related Apoptosis-Inducing Ligand/pharmacology , Antineoplastic Agents/pharmacology , BH3 Interacting Domain Death Agonist Protein/metabolism , Caspases/metabolism , Cell Line, Tumor , Cell Survival/drug effects , Down-Regulation/drug effects , Drug Synergism , Humans , Membrane Potential, Mitochondrial/drug effects , Myeloid Cell Leukemia Sequence 1 Protein , Neoplasm Proteins/metabolism , Proto-Oncogene Proteins c-bcl-2/metabolism , Transduction, Genetic , Vorinostat , X-Linked Inhibitor of Apoptosis Protein/genetics , bcl-X Protein/metabolism
5.
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
6.
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
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.
Cancer Gene Ther ; 12(11): 864-72, 2005 Nov.
Article in English | MEDLINE | ID: mdl-15891771

ABSTRACT

Current treatment regimens for patients with metastatic melanoma are not curative, and new treatment strategies are needed. One possible approach is targeted treatment using the tyrosinase promoter for melanoma-specific expression of genes delivered by adenoviral (Ad) vectors. In this study, a vector with the human minimal tyrosinase promoter and two human enhancer elements (2hE-hTyrP) was compared with different hybrid promoter constructs, containing tyrosinase regulatory sequences and the viral simian virus 40 (SV40) promoter. The tissue specificity of the first-generation vectors was measured by enhanced green fluorescence protein (EGFP) reporter flow cytometry in 12 human melanoma and nonmelanoma cell lines. In the melanotic melanoma cells, the activity of the 2hE-hTyrP promoter was comparable with the activity of the cytomegalovirus promoter, and the background expression levels obtained in the nonmelanoma cell lines confirmed the strict tissue-specific property of this promoter. The hybrid SV40-based promoters were effective, but no tissue specificity was observed even after the inclusion of tyrosinase enhancer elements identical to the elements used in the 2hE-hTyrP promoter. The in vivo tissue specificity of the 2hE-hTyrP vector was demonstrated in subcutaneous xenografted tumors by ex vivo detection of EGFP fluorescence with the IVIS Imaging equipment and fluorescence microscopy visualizing the in situ EGFP expression in tumor sections. The tyrosinase mRNA level in the 12 cell lines was measured by quantitative real-time RT-PCR, and the expression levels reliably reflected to what extent the 2hE-hTyrP promoter could drive the gene expression in the individual cell lines. In conclusion, the human tyrosinase promoter fused to two human tyrosinase enhancers (2hE-hTyrP) can be used for efficient tissue-specific expression from first-generation Ad vectors in melanoma cell lines both in vitro and in vivo, as predicted by the quantitative tyrosinase mRNA levels in the melanoma and nonmelanoma cell lines tested.


Subject(s)
Genetic Therapy/methods , Melanoma/genetics , Melanoma/therapy , Monophenol Monooxygenase/genetics , Skin Neoplasms/genetics , Skin Neoplasms/therapy , Adenoviridae/genetics , Enhancer Elements, Genetic , Flow Cytometry , Gene Expression Profiling , Genes, Reporter , Genetic Vectors , Green Fluorescent Proteins/analysis , Humans , Monophenol Monooxygenase/biosynthesis , Promoter Regions, Genetic , Simian virus 40/genetics , Tissue Distribution , Transfection , Tumor Cells, Cultured
9.
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
10.
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
11.
Photochem Photobiol Sci ; 2(4): 407-11, 2003 Apr.
Article in English | MEDLINE | ID: mdl-12760539

ABSTRACT

Entrapment and degradation of transfecting DNA in endocytic vesicles often hampers the use of lipidic vectors for gene delivery purposes. Photochemical internalisation (PCI) is a technology for achieving light-induced release of DNA trapped inside these vesicles, and therefore represents a way of overcoming the endocytic membrane barrier and improving gene transfer. The technology is based on utilising photosensitizers which localise in the membranes of endocytic vesicles, causing photochemical damages that rupture the vesicles upon illumination. The purpose of this work was to study the effect of PCI on transfection mediated by the cationic lipid N-(2-aminoethyl)-N,N-dimethyl-2,3-bis(tetradecyloxy)-1-propanaminium bromide (betaAE-DMRIE), with or without the helper lipid 1,2-dioleoyl-sn-glycero-3-phosphoethanolamine (DOPE). It was shown that PCI has no effect on betaAE-DMRIE mediated transfection, whereas it significantly enhances transfection mediated by the combination of betaAE-DMRIE and DOPE. The effect of PCI was highly dependent on the timing of illumination relative to the time of DNA delivery, both regarding the sequence of, and the time between, these two treatments.


Subject(s)
DNA/administration & dosage , Lipids/administration & dosage , Photochemistry , Transfection/methods , Cations , Cell Survival , Flow Cytometry , Humans , Tumor Cells, Cultured
12.
Somat Cell Mol Genet ; 27(1-6): 97-113, 2002 Nov.
Article in English | MEDLINE | ID: mdl-12774944

ABSTRACT

Most synthetic gene delivery vectors are taken up in the cell by endocytosis, and inefficient escape of the transgene from endocytic vesicles often is a major barrier for gene transfer by such vectors. To improve endosomal release we have developed a new technology, named photochemical internalization (PCI). PCI is based on photochemical reactions initiated by photosensitizing compounds localized in endocytic vesicles, inducing rupture of these vesicles upon light exposure. PCI constitutes an efficient light-inducible gene transfer method in vitro, which potentially can be developed into a site-specific method for gene delivery in in vivo gene therapy. In this paper the principle behind the PCI technology and the effect of PCI on transfection with different synthetic gene delivery vectors are reviewed. PCI treatment by the photosensitizer aluminum phthalocyanine (AlPcS2a) strongly improves transfection mediated by cationic polymers (e.g., poly-L-lysine and polyethylenimine), while the effect on transfection with cationic lipids is more variable. The timing of the light treatment relative to the transfection period was also important, indicating that release of the DNA from early endosomes is important for the outcome of PCI-induced transfection. The possibilities of using PCI as a technology for efficient, site-specific gene delivery in in vivo gene therapy is discussed.


Subject(s)
Gene Transfer Techniques , Photochemistry/methods , Transfection/methods , Animals , Cell Line , DNA/administration & dosage , DNA/chemistry , Drug Delivery Systems , Endocytosis/physiology , Gene Transfer Techniques/trends , Genetic Therapy/methods , Genetic Therapy/trends , Genetic Vectors/administration & dosage , Genetic Vectors/genetics , Humans , Light , Neoplasms/therapy , Photosensitizing Agents/chemistry , Photosensitizing Agents/pharmacokinetics , Photosensitizing Agents/pharmacology , Transcription, Genetic , Transfection/trends
SELECTION OF CITATIONS
SEARCH DETAIL
...