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










Publication year range
1.
Oncologist ; 27(6): 430-e433, 2022 06 08.
Article in English | MEDLINE | ID: mdl-35675633

ABSTRACT

BACKGROUND: Photochemical internalization (PCI) is a novel technology for light-induced enhancement of the local therapeutic effect of cancer drugs, utilizing a specially designed photosensitizing molecule (fimaporfin). The photosensitizing molecules are trapped in endosomes along with macromolecules or drugs. Photoactivation of fimaporfin disrupts the endosomal membranes so that drug molecules are released from endosomes inside cells and can reach their therapeutic target in the cell cytosol or nucleus. Compared with photodynamic therapy, the main cytotoxic effect with PCI is disruption of the endosomal membrane resulting in delivery of chemotherapy drug, and not to the photochemical reactions per se. In this study we investigated the effect of PCI with gemcitabine in patients with inoperable perihilar cholangiocarcinoma (CCA). METHODS: The in vitro cytotoxic effect of PCI with gemcitabine was studied on two CCA-derived cell lines. In a fimaporfin dose-escalation phase I clinical study, we administered PCI with gemcitabine in patients with perihilar CCA (n = 16) to establish a safe and tolerable fimaporfin dose and to get early signals of efficacy. The patients enrolled in the study had tumors in which the whole length of the tumor could be illuminated from the inside of the bile duct, using an optical fiber inserted via an endoscope (Fig. 1). Fimaporfin was administered intravenously at day 0; gemcitabine (i.v.) and intraluminal biliary endoscopic laser light application on day 4; followed by standard gemcitabine/cisplatin chemotherapy. RESULTS: Preclinical experiments showed that PCI enhanced the effect of gemcitabine. In patients with CCA, PCI with gemcitabine was well tolerated with no dose-limiting toxicities, and no unexpected safety signals. Disease control was achieved in 10 of 11 evaluable patients, with a clearly superior effect in the two highest dose groups. The objective response rate (ORR) was 42%, including two complete responses, while ORR at the highest dose was 60%. Progression-free survival at 6 months was 75%, and median overall survival (mOS) was 15.4 months, with 22.8 months at the highest fimaporfin dose. CONCLUSION: Photochemical internalization with gemcitabine was found to be safe and resulted in encouraging response and survival rates in patients with unresectable perihilar CCA.


Subject(s)
Cholangiocarcinoma , Deoxycytidine , Photochemotherapy , Antineoplastic Agents/therapeutic use , Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Bile Duct Neoplasms/drug therapy , Bile Ducts, Intrahepatic , Cholangiocarcinoma/drug therapy , Cholangiocarcinoma/pathology , Deoxycytidine/adverse effects , Deoxycytidine/analogs & derivatives , Humans , Photochemotherapy/adverse effects , Photochemotherapy/methods , Gemcitabine
2.
Cells ; 10(6)2021 06 12.
Article in English | MEDLINE | ID: mdl-34204611

ABSTRACT

Fibroblast growth factor receptors (FGFRs) have become an attractive target in cancer research and therapy due to their implication in several cancers. Limitations of current treatment options require a need for additional, more specific and potent strategies to overcome cancers driven by FGFRs. Photochemical internalization (PCI) is a light-controlled method for cytosolic delivery of drugs that are entrapped in endosomes and lysosomes. We here evaluated the efficacy and selectivity of PCI of FGF2-saporin (FGF-SAP) in cells overexpressing FGFR1. FGF-SAP is a conjugate of FGF2 and the highly cytotoxic ribosome-inactivating protein (RIP) saporin, which is used as payload to eliminate cancer cells. Evaluation of the targeting effect of PCI of FGF-SAP was done by comparing the cytotoxic response in osteosarcoma cells with very low levels of FGFR1 (U2OS) to cells overexpressing FGFR1 (U2OS-R1). We demonstrate that PCI greatly enhances cytotoxicity of the drug showing efficient cell killing at pM concentrations of the drug in U2OS-R1 cells. However, U2OS cells were also sensitive to the toxin after PCI. Binding experiments using confocal microscopy and Western blotting techniques indicate that FGF-SAP is taken up by cells through heparan sulfate proteoglycans (HSPGs) in U2OS cells. We further show that the cytotoxicity of FGF-SAP in U2OS cells was reduced when cells were co-treated with heparin to compete out binding to HSPG, demonstrating that the cytotoxic effect was due to internalization by HSPGs. We conclude that to prevent off-target effects of FGF-based toxins, it will be necessary to circumvent binding to HSPGs, for example by mutating the binding site of FGF2 to HSPGs.


Subject(s)
Drug Carriers , Fibroblast Growth Factor 2 , Molecular Targeted Therapy/methods , Photochemotherapy/methods , Saporins/administration & dosage , Cell Line, Tumor , Drug Delivery Systems/methods , Humans
3.
Front Immunol ; 9: 650, 2018.
Article in English | MEDLINE | ID: mdl-29670624

ABSTRACT

Effective priming and activation of tumor-specific CD8+ cytotoxic T lymphocytes (CTLs) is crucial for realizing the potential of therapeutic cancer vaccination. This requires cytosolic antigens that feed into the MHC class I presentation pathway, which is not efficiently achieved with most current vaccination technologies. Photochemical internalization (PCI) provides an emerging technology to route endocytosed material to the cytosol of cells, based on light-induced disruption of endosomal membranes using a photosensitizing compound. Here, we investigated the potential of PCI as a novel, minimally invasive, and well-tolerated vaccination technology to induce priming of cancer-specific CTL responses to peptide antigens. We show that PCI effectively promotes delivery of peptide antigens to the cytosol of antigen-presenting cells (APCs) in vitro. This resulted in a 30-fold increase in MHC class I/peptide complex formation and surface presentation, and a subsequent 30- to 100-fold more efficient activation of antigen-specific CTLs compared to using the peptide alone. The effect was found to be highly dependent on the dose of the PCI treatment, where optimal doses promoted maturation of immature dendritic cells, thus also providing an adjuvant effect. The effect of PCI was confirmed in vivo by the successful induction of antigen-specific CTL responses to cancer antigens in C57BL/6 mice following intradermal peptide vaccination using PCI technology. We thus show new and strong evidence that PCI technology holds great potential as a novel strategy for improving the outcome of peptide vaccines aimed at triggering cancer-specific CD8+ CTL responses.


Subject(s)
CD8-Positive T-Lymphocytes/immunology , Cancer Vaccines/immunology , Immunotherapy/methods , Neoplasms/therapy , Vaccination/methods , Animals , Antigen Presentation , Antigens, Neoplasm/immunology , Cytotoxicity, Immunologic , Endocytosis , Humans , Injections, Intradermal , Mice , Neoplasms/immunology , Peptides/immunology , Photochemical Processes
4.
Photochem Photobiol Sci ; 14(8): 1465-75, 2015 Aug.
Article in English | MEDLINE | ID: mdl-25849953

ABSTRACT

Resistance to chemotherapy, molecular targeted therapy as well as radiation therapy is a major obstacle for cancer treatment. Cancer resistance may be exerted through multiple different mechanisms which may be orchestrated as observed in multidrug resistance (MDR). Cancer resistance may be intrinsic or acquired and often leaves patients without any treatment options. Strategies for alternative treatment modalities for resistant cancer are therefore highly warranted. Photochemical internalization (PCI) is a technology for cytosolic delivery of macromolecular therapeutics based on the principles of photodynamic therapy (PDT). The present report reviews the current knowledge of PCI of therapy-resistant cancers. In summary, PCI may be able to circumvent several of the major mechanisms associated with resistance towards chemotherapeutics including increased expression of drug efflux pumps, altered intracellular drug distribution and increased ROS scavenging. Current data also suggest PCI of targeted toxins as highly effective in cancers resistant to clinically available targeted therapy such as monoclonal antibodies (mAbs) and tyrosine kinase inhibitors (TKIs). PCI may therefore, in general, represent a future treatment option for cancers resistant to other therapies.


Subject(s)
Drug Resistance, Neoplasm , Neoplasms/drug therapy , Photochemotherapy/methods , Photosensitizing Agents/administration & dosage , Humans , Neoplasms/physiopathology
5.
J Control Release ; 198: 10-7, 2015 Jan 28.
Article in English | MEDLINE | ID: mdl-25482339

ABSTRACT

Cancer vaccines aim to induce CD8 T cells infiltrating the tumour. For protein-based vaccines, the main biological barrier to overcome is the default MHC class-II-pathway, with activation of CD4 T cells rather than CD8 T cells. The latter requires antigens to access the cytosol and MHC class I antigen presentation. We applied photosensitiser and light to trigger disruption of antigen-containing endosomes and thereby MHC class I cross-presentation of a model cancer vaccine. This "photochemical internalisation" resulted in activation, proliferation, and IFN-γ production of cytotoxic CD8 T cells, which suppressed tumour growth by infiltrating CD8 T cells and caspase-3-dependent apoptosis. The process was independent of MHC class II, MyD88, and TLR4 signalling, but dependent on trypsin- and caspase-like proteasome activity and partly also on chloroquine. This novel method of vaccination may find applications in cancer immunotherapy where the activation of CD8 T cells is important.


Subject(s)
CD8-Positive T-Lymphocytes/immunology , Cancer Vaccines , Melanoma/therapy , Photosensitizing Agents/therapeutic use , Porphyrins/therapeutic use , Skin Neoplasms/therapy , Animals , Cell Proliferation , Cross-Priming , Dendritic Cells/immunology , Histocompatibility Antigens Class I/immunology , Interferon-gamma/immunology , Light , Melanoma/immunology , Mice, Inbred C57BL , Mice, Transgenic , Ovalbumin/immunology , Photosensitivity Disorders , Skin Neoplasms/immunology , Spleen/cytology
6.
MAbs ; 6(4): 1038-50, 2014.
Article in English | MEDLINE | ID: mdl-24525727

ABSTRACT

The epithelial cell adhesion molecule (EpCAM) is expressed by a wide range of human carcinomas, making it an attractive diagnostic and therapeutic target in oncology. Its recent identification on cancer stem cells has raised further interest in its use for tumor targeting and therapy. Here, we present the characterization and therapeutic potential of 3-17I, a novel human EpCAM-targeting monoclonal antibody. Strong reaction of 3-17I was observed in all lung, colon, and breast human tumor biopsies evaluated. By flow cytometry and confocal fluorescence microscopy, we demonstrate that 3-17I specifically targets EpCAM-positive cell lines. We also show evidence for mAb-sequestration in endo-/lysosomes, suggesting internalization of 3-17I by receptor-mediated endocytosis. The ribosomal-inactivating toxin saporin was linked to 3-17I, creating the per se non-toxic immunotoxin 3-17I-saporin, a promising candidate for the drug delivery technology photochemical internalization (PCI). PCI is based on a light-controlled destruction of endolysosomal membranes and subsequent cytosolic release of the sequestered payload upon light exposure. EpCAM-positive human cancer cell lines MCF7 (breast), BxPC-3 (pancreas), WiDr (colon), and the EpCAM-negative COLO320DM (colon), were treated with 3-17I-saporin in combination with the clinically relevant photosensitizer TPCS2a (Amphinex), followed by exposure to light. No cytotoxicity was observed after treatment with 3-17I-saporin without light exposure. However, cell viability, proliferation and colony-forming capacity was strongly reduced in a light-dependent manner after PCI of 3-17I. Our results show that 3-17I is an excellent candidate for diagnosis of EpCAM-positive tumors and for development of clinically relevant antibody-drug conjugates, using PCI for the treatment of localized tumors.


Subject(s)
Antibodies, Monoclonal, Murine-Derived/pharmacology , Antibodies, Neoplasm/pharmacology , Breast Neoplasms/drug therapy , Colonic Neoplasms/drug therapy , Immunotoxins/pharmacology , Pancreatic Neoplasms/drug therapy , Ribosome Inactivating Proteins, Type 1/pharmacology , Animals , Antibodies, Monoclonal, Murine-Derived/immunology , Antibodies, Neoplasm/immunology , Breast Neoplasms/immunology , Breast Neoplasms/pathology , Cell Line, Tumor , Colonic Neoplasms/immunology , Colonic Neoplasms/pathology , Drug Delivery Systems/methods , Female , Humans , Immunotoxins/immunology , Pancreatic Neoplasms/immunology , Pancreatic Neoplasms/pathology , Photochemistry/methods , Ribosome Inactivating Proteins, Type 1/immunology , Saporins
7.
Biochim Biophys Acta ; 1830(3): 2659-70, 2013 Mar.
Article in English | MEDLINE | ID: mdl-23671927

ABSTRACT

BACKGROUND: Tyrosin kinase inhibitors (TKIs) and monoclonal antibodies aimed to target epidermal growth factor receptor (EGFR) have shown limited effect as monotherapies and drug resistance is a major limitation for therapeutic success. Adjuvant therapies to EGFR targeting therapeutics are therefore of high clinical relevance. METHODS: Three EGFR targeting drugs, Cetuximab, Erlotinib and Tyrphostin AG1478 were used in combination with photodynamic therapy (PDT) in two EGFR positive cell lines, A-431 epidermoid skin carcinoma and WiDr colorectal adenocarcinoma cells. The amphiphilic meso-tetraphenylporphine with 2 sulphonate groups on adjacent phenyl rings (TPPS(2a)) was utilized as a photosensitizer for PDT. The cytotoxic outcome of the combined treatments was evaluated by cell counting and MTT. Cellular signalling was explored by Western blotting. RESULTS: PDT as neoadjuvant to Tyrphostin in A-431 cells as well as to Tyrphostin or Erlotinib in WiDr cells revealed synergistic cytotoxicity. In contrast, Erlotinib or Cetuximab combined with neoadjuvant PDT induced an antagonistic effect on cell survival of A-431 cells. Neoadjuvant PDT and EGFR targeting therapies induced a synergistic inhibition of ERK as well as synergistic cytotoxicity only when the EGFR targeting monotherapies caused a prolonged ERK inhibition. There were no correlation between EGFR inhibition by the EGFR targeting monotherapies or the combined therapies and the cytotoxic outcome combination-therapies. CONCLUSIONS: The results suggest that sustained ERK inhibition by EGFR targeting monotherapies is a predictive factor for synergistic cytotoxicity when combined with neoadjuvant PDT. GENERAL SIGNIFICANCE: The present study provides a rationale for selecting anticancer drugs which may benefit from PDT as adjuvant therapy.


Subject(s)
Antineoplastic Agents/pharmacology , ErbB Receptors/genetics , Extracellular Signal-Regulated MAP Kinases/antagonists & inhibitors , Photosensitizing Agents/pharmacology , Antibodies, Monoclonal, Humanized/pharmacology , Cell Count , Cell Line, Tumor/drug effects , Cell Survival/drug effects , Cetuximab , Drug Synergism , ErbB Receptors/metabolism , Erlotinib Hydrochloride , Extracellular Signal-Regulated MAP Kinases/genetics , Extracellular Signal-Regulated MAP Kinases/metabolism , Gene Expression Regulation/drug effects , Humans , Molecular Targeted Therapy , Neoadjuvant Therapy , Photochemotherapy , Porphyrins/pharmacology , Quinazolines/pharmacology , Signal Transduction/drug effects , Tyrphostins/pharmacology
8.
J Control Release ; 168(3): 317-26, 2013 Jun 28.
Article in English | MEDLINE | ID: mdl-23567040

ABSTRACT

CD133 is a putative cancer stem cell (CSC) marker for a number of different cancers and is suggested to be a therapeutic target. Since also normal stem cells express CD133 it is of paramount importance that targeting strategies provide a specific and efficient delivery of cytotoxic drugs in only CD133-positive CSCs. In this study, we have employed photochemical internalization (PCI), a minimally invasive method for light-controlled, specific delivery of membrane-impermeable macromolecules from endocytic vesicles to the cytosol, to specifically target CD133-positive cancer cells. We demonstrate that PCI increases the cytotoxic effect of an immunotoxin (IT) targeting CD133-expressing cancer cells of colon (WiDr and HCT116) and pancreas (BxPC-3) origin. The IT consisted of the mAb CD133/1 (AC133) bound to the ribosome inactivating plant toxin saporin (anti-CD133/1-sap). We show that TPCS2a-PCI of anti-CD133/1-sap is specific, and highly cytotoxic at femto-molar concentrations. Specific binding and uptake of CD133/1, was shown by fluorescence microscopy and co-localization with TPCS2a in endosomes/lysosomes was determined by confocal microscopy. CD133(high) WiDr cells, isolated by fluorescence activated cell sorting, had a 7-fold higher capacity to initiate spheroids than CD133(low) cells (P<0.001) and were resistant to photodynamic therapy (PDT). However, PDT-resistance was bypassed by the PCI strategy. Tumor initiation and aggressive growth in athymic nude mice was obtained with only 10 CD133(high) cells in contrast to CD133(low) cells where substantially higher cell numbers were needed. The excellent high efficacy and selectivity of eliminating CD133-expressing cells by PCI warrant further pre-clinical evaluations of this novel therapeutic approach.


Subject(s)
Antibodies, Monoclonal/administration & dosage , Antigens, CD/immunology , Glycoproteins/immunology , Neoplastic Stem Cells/immunology , Peptides/immunology , Photosensitizing Agents/administration & dosage , Porphyrins/administration & dosage , Ribosome Inactivating Proteins, Type 1/administration & dosage , AC133 Antigen , Antibodies, Monoclonal/chemistry , Cell Line, Tumor , Drug Delivery Systems , Humans , Photochemical Processes , Photochemotherapy , Ribosome Inactivating Proteins, Type 1/chemistry , Saporins
9.
Cancer Lett ; 321(2): 120-7, 2012 Aug 28.
Article in English | MEDLINE | ID: mdl-22266098

ABSTRACT

Targeted photosensitizer delivery to EGFR-expressing cells was achieved in the present study using a high purity, targeted photoimmunoconjugate (PIC). When the PDT agent, benzoporphyrin derivative monoacid ring A (BPD) was coupled to an EGFR-targeting antibody (cetuximab), we observed altered cellular localization and selective phototoxicity of EGFR-positive cells, but no phototoxicity of EGFR-negative cells. Cetuximab in the PIC formulation blocked EGF-induced activation of the EGFR and downstream signaling pathways. Our results suggest that photoimmunotargeting is a useful dual strategy for the selective destruction of cancer cells and also exerts the receptor-blocking biological function of the antibody.


Subject(s)
Antibodies, Monoclonal/pharmacology , Antineoplastic Agents/pharmacology , Epidermal Growth Factor/metabolism , ErbB Receptors/antagonists & inhibitors , Ovarian Neoplasms/drug therapy , Photochemotherapy/methods , Photosensitizing Agents/pharmacology , Porphyrins/pharmacology , Signal Transduction/drug effects , Antibodies, Monoclonal, Humanized , Blotting, Western , Cell Line, Tumor , Cetuximab , Dermatitis, Phototoxic , Epidermal Growth Factor/pharmacology , ErbB Receptors/immunology , ErbB Receptors/metabolism , Female , Humans , Signal Transduction/physiology , Verteporfin
10.
Lasers Surg Med ; 43(7): 721-33, 2011 Sep.
Article in English | MEDLINE | ID: mdl-22057500

ABSTRACT

Photochemical internalization (PCI) is a method for intracellular delivery of hydrophilic macromolecular drugs with intracellular targets as well as other drugs with limited ability to penetrate cellular membranes. Such drugs enter cells by means of endocytosis and are to a large extent degraded by hydrolytic enzymes in the lysosomes unless they possess a mechanism for cytosolic translocation. PCI is based on photodynamic therapy (PDT) specifically targeting the endosomes and lysosomes of the cells, so that the drugs in these vesicles can escape into the cytosol from where they can reach their targets. The preferential retention of the photosensitizer (PS) in tumor tissue in combination with controlled light delivery makes PCI relatively selective for cancer tissue. The tumor specificity of PCI can be further increased by delivery of drugs that selectively target the tumors. Indeed, this has been shown by PCI delivery of several targeted protein toxins. Targeted protein toxins may be regarded as ideal drugs for PCI delivery, and may represent the clinical future for the PCI technology.


Subject(s)
Antineoplastic Agents/administration & dosage , Immunotoxins/administration & dosage , Molecular Targeted Therapy/methods , Neoplasms/drug therapy , Photochemotherapy , Antineoplastic Agents/pharmacokinetics , Antineoplastic Agents/therapeutic use , Drug Delivery Systems , Endosomes/drug effects , Humans , Immunotoxins/pharmacokinetics , Immunotoxins/therapeutic use , Lysosomes/drug effects , Macromolecular Substances/administration & dosage , Macromolecular Substances/pharmacokinetics , Macromolecular Substances/therapeutic use , Photosensitizing Agents/pharmacology
11.
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
12.
J Control Release ; 142(3): 347-53, 2010 Mar 19.
Article in English | MEDLINE | ID: mdl-19932724

ABSTRACT

Photochemical internalisation (PCI) is a delivery technology that employs a sub-lethal form of photodynamic therapy (PDT) in which a photosensitiser is activated by light to break down intracellular membranes and release macromolecules into the cytosol where they can be biologically active. Although PCI does enhance the PDT killing of transplanted tumours in mice after local injection of the cytotoxic agent, gelonin, the redistribution of gelonin from intracellular organelles into the cytosol has only previously been demonstrated in vitro. This study is designed to understand the factors controlling the efficacy of PCI in vivo and to document the mechanism of action. Using the photosensitiser AlS(2)Pc in studies on normal rat liver, we have demonstrated in vivo that gelonin is initially taken up into lysosomes, but can be released into the cytosol using PCI. Furthermore, PCI enhances the PDT effect after systemic administration of gelonin (volume of necrosis increased x2.5 when gelonin is given one hour before light), with the remarkably low dose of 5 microg/kg (10,000 times lower than the LD50); in the absence of light, there is no effect with 500 microg/kg. These results suggest that PCI may have a useful role to play in the site specific activation of cytotoxic agents like gelonin, given at a dose level that has no effect in the absence of light.


Subject(s)
Antineoplastic Agents, Phytogenic/pharmacology , Indoles/pharmacology , Liver/drug effects , Organometallic Compounds/pharmacology , Photochemotherapy/methods , Photosensitizing Agents/pharmacology , Ribosome Inactivating Proteins, Type 1/pharmacology , Animals , Antineoplastic Agents, Phytogenic/administration & dosage , Antineoplastic Agents, Phytogenic/pharmacokinetics , Antineoplastic Agents, Phytogenic/therapeutic use , Cytosol/metabolism , Dose-Response Relationship, Drug , Drug Administration Schedule , Female , Indoles/administration & dosage , Indoles/pharmacokinetics , Indoles/therapeutic use , Light , Liver/metabolism , Liver/pathology , Liver/radiation effects , Lysosomes/metabolism , Necrosis , Organometallic Compounds/administration & dosage , Organometallic Compounds/pharmacokinetics , Organometallic Compounds/therapeutic use , Photochemical Processes , Photosensitizing Agents/administration & dosage , Photosensitizing Agents/pharmacokinetics , Photosensitizing Agents/therapeutic use , Rats , Rats, Wistar , Ribosome Inactivating Proteins, Type 1/administration & dosage , Ribosome Inactivating Proteins, Type 1/pharmacokinetics , Ribosome Inactivating Proteins, Type 1/therapeutic use
13.
Top Curr Chem ; 296: 251-81, 2010.
Article in English | MEDLINE | ID: mdl-21504105

ABSTRACT

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. Upon activation by light such photosensitizers induce a release of macromolecules from their compartmentalization in endocytic vesicles. PCI has been shown to increase 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, immunotoxins, plasmids, adenovirus, various oligonucleotides, dendrimer-based delivery of chemotherapeutica and unconjugated chemotherapeutica such as bleomycin and doxorubicin. This review will present the basis for the PCI concept and the most recent significant developments.


Subject(s)
Oligonucleotides/genetics , Photochemical Processes , Photosensitizing Agents/metabolism , Transfection/methods , Cytosol/drug effects , Cytosol/metabolism , Endocytosis/drug effects , Oligonucleotides/metabolism , Photosensitizing Agents/chemistry , Photosensitizing Agents/pharmacology
14.
PLoS One ; 4(8): e6691, 2009 Aug 19.
Article in English | MEDLINE | ID: mdl-19690617

ABSTRACT

BACKGROUND: There is a need for drug delivery systems (DDS) that can enhance cytosolic delivery of anti-cancer drugs trapped in the endo-lysosomal compartments. Exposure of cells to specific photosensitizers followed by light exposure (photochemical internalization, PCI) results in transfer of agents from the endocytic compartment into the cytosol. METHODOLOGY AND PRINCIPAL FINDINGS: The recombinant single-chain fusion construct scFvMEL/rGel is composed of an antibody targeting the progenitor marker HMW-MAA/NG2/MGP/gp240 and the highly effective toxin gelonin (rGel). Here we demonstrate enhanced tumor cell selectivity, cytosolic delivery and anti-tumor activity by applying PCI of scFvMEL/rGel. PCI performed by light activation of cells co-incubated with scFvMEL/rGel and the endo-lysosomal targeting photosensitizers AlPcS(2a) or TPPS(2a) resulted in enhanced cytotoxic effects against antigen-positive cell lines, while no differences in cytotoxicity between the scFvMEL/rGel and rGel were observed in antigen-negative cells. Mice bearing well-developed melanoma (A-375) xenografts (50-100 mm(3)) were treated with PCI of scFvMEL/rGel. By 30 days after injection, approximately 100% of mice in the control groups had tumors>800 mm(3). In contrast, by day 40, 50% of mice in the PCI of scFvMEL/rGel combination group had tumors<800 mm(3) with no increase in tumor size up to 110 days. PCI of scFvMEL/rGel resulted in a synergistic effect (p<0.05) and complete regression (CR) in 33% of tumor-bearing mice (n = 12). CONCLUSIONS/SIGNIFICANCE: This is a unique demonstration that a non-invasive multi-modality approach combining a recombinant, targeted therapeutic such as scFvMEL/rGel and PCI act in concert to provide potent in vivo efficacy without sacrificing selectivity or enhancing toxicity. The present DDS warrants further evaluation of its clinical potential.


Subject(s)
Antineoplastic Agents/administration & dosage , Drug Delivery Systems , Immunoglobulin Fragments/administration & dosage , Immunotoxins/administration & dosage , Recombinant Fusion Proteins/administration & dosage , Animals , Antineoplastic Agents/therapeutic use , Cell Line, Tumor , Female , Humans , Immunotoxins/therapeutic use , Mice , Mice, Inbred BALB C , Photochemistry , Recombinant Fusion Proteins/therapeutic use
15.
Mol Pharm ; 6(1): 255-64, 2009.
Article in English | MEDLINE | ID: mdl-19125612

ABSTRACT

Mammalian target of rapamycin (mTOR) is a regulator of cell growth and proliferation and its activity is altered in many human cancers. The main objective of this study was to evaluate in vitro and in vivo targeting of mTOR by photodynamic therapy (PDT), a treatment modality for cancer. The amphiphilic endolysosomal localizing photosensitizer AlPcS(2a) and the p53 mutated rapamycin-resistant colon adenocarcinoma cell line WiDr were used as models. AlPcS(2a)-PDT downregulated the levels of Ser(2448) phosphorylated mTOR (p-mTOR), total mTOR and phosphorylation of ribosomal S6 (p-S6) immediately after light exposure in a dose-dependent manner, indicating a direct targeting of the mTOR signaling network. Low-dose PDT attenuated the level of p-mTOR in a transient manner; approximately 35% reduction of p-mTOR was obtained 5 min after a LD(35) PDT dose, but returned to the basal level 24 h later. Treatment with the mTOR inhibitor rapamycin reduced the p-mTOR level by 25% after 4-24 h of incubation. Combination treatment of rapamycin and PDT in vitro resulted in synergistic cytotoxic effects when rapamycin was administered after PDT. However, antagonistic effects were obtained when rapamycin was incubated both before and after PDT. In vivo, activated mTOR in the WiDr-xenografts was downregulated by 35 and 75% 5 min and 24 h post PDT respectively as measured by immunoblotting. In contrast to untreated tumors where p-mTOR expression was found throughout the tumors, immunohistochemical staining revealed only expression of p-mTOR in the rim of the tumor at 24 and 48 h post PDT. In conclusion, AlPcS(2a)-PDT is a novel mTOR-targeted cancer therapy. Rapamycin synergistically enhances the cytotoxicity of PDT only when administered post light exposure.


Subject(s)
Protein Kinases/metabolism , Signal Transduction/drug effects , Animals , Cell Line, Tumor , Drug Synergism , Female , Humans , Mice , Mice, Inbred BALB C , Mice, Nude , Phosphorylation/drug effects , Photochemotherapy , Sirolimus/metabolism , TOR Serine-Threonine Kinases
16.
Cancer Biother Radiopharm ; 22(4): 469-79, 2007 Aug.
Article in English | MEDLINE | ID: mdl-17803441

ABSTRACT

Radioimmunotherapy (RIT) with the alpha-emitter 227Th is currently under evaluation. 227Th is conjugated to the chimeric anti-CD20 monoclonal antibody rituximab, using the chelator p-isothiocyanato-benzyl-DOTA. In this study, the binding of 227Th-DOTA-p-benzyl-rituximab to three different CD-20-positive lymphoma cell lines, Raji, Rael, and Daudi, were evaluated. Equilibrium and kinetic binding experiments were used to determine binding parameters, including the association and dissociation rate constants, the equilibrium dissociation constants, and the total number of antigens for Raji, Rael, and Daudi cells. There were significant differences between the cell lines with respect to both Kd and the total number of antigens. Rael cells had more than three times as many antigens as the other two cell lines, and the functional Kd found for Rael cells was significantly higher than that found for Raji and Daudi cells. These results were confirmed using flow cytometry. Rituximab was found to be localized in patches on the cell membrane. The findings indicated that 227Th-labeled rituximab has relevant antigen-targeting properties for radioimmunotherapy.


Subject(s)
Antibodies, Monoclonal/immunology , Antibodies, Monoclonal/pharmacology , Antigens, CD20/immunology , Lymphoma/immunology , Thorium/chemistry , Thorium/pharmacology , Antibodies, Monoclonal/chemistry , Antibodies, Monoclonal, Murine-Derived , Antigens, CD20/metabolism , Cell Line, Tumor , Feasibility Studies , Humans , Kinetics , Lymphoma/metabolism , Lymphoma/pathology , Radioimmunotherapy , Rituximab
17.
Mol Pharm ; 4(2): 241-51, 2007.
Article in English | MEDLINE | ID: mdl-17263556

ABSTRACT

Photochemical internalization (PCI) is a novel technology of macromolecular delivery. By PCI, endocytosed membrane-impermeable therapeutic drugs are photochemically released from entrapment in endo-lysosomal compartments to the cytosol of target cells. In the present report, we describe the in vitro proof-of-concept for PCI of cetuximab-saporin, an immunotoxin targeting EGFR-expressing cells. This immunotoxin consists of the chimeric murine-human IgG1 monoclonal antibody cetuximab (C225 or Erbitux) bound to the type I ribosome-inactivating protein toxin saporin by a biotin-streptavidin linkage. The photochemical treatment enhanced the cytotoxicity of the immunotoxin in a synergistic manner in three different EGFR-expressing carcinoma cell lines derived from different tumor tissues (colorectal, HCT-116; prostate, DU-145; and epidermis, A-431). Both cytotoxicity of cetuximab-saporin and epifluorescence of Alexa488-cetuximab were evaluated by competition with cetuximab demonstrating specific binding and uptake of cetuximab-saporin in EGFR positive cells. In the EGFR-negative uterine sarcoma cell line MES-SA, neither binding nor preferential accumulation of Alexa488-cetuximab was detected. In addition, PCI enhanced the cytotoxicity of cetuximab-saporin to the same extent as streptavidin-saporin in the MES-SA cells. In conclusion, PCI enhances selectivity of the cytotoxicity of the immunotoxin cetuximab-saporin in EGFR-expressing cells.


Subject(s)
Antibodies, Monoclonal , Drug Delivery Systems/methods , ErbB Receptors/metabolism , Neoplasms/drug therapy , Saponins/pharmacology , Antibodies, Monoclonal/pharmacokinetics , Antibodies, Monoclonal/pharmacology , Antibodies, Monoclonal, Humanized , Cell Line, Tumor , Cell Survival , Cetuximab , Dose-Response Relationship, Drug , Drug Synergism , Humans , Light , Models, Biological , Phosphorylation , Photochemotherapy
18.
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
19.
Cancer Res ; 66(11): 5633-40, 2006 Jun 01.
Article in English | MEDLINE | ID: mdl-16740700

ABSTRACT

Photodynamic therapy (PDT) is now an approved therapeutic modality, and induction of vascular endothelial growth factor (VEGF) following subcurative PDT is of concern as VEGF may provide a survival stimulus to tumors. The processes that limit the efficacy of PDT warrant investigation so that mechanism-based interventions may be developed. This study investigates VEGF increase following subcurative PDT using the photosensitizer benzoporphyrin derivative (BPD) both in an in vitro and in an orthotopic model of prostate cancer using the human prostate cancer cell line LNCaP. The two subcurative doses used, 0.25 and 0.5 J/cm(2), mimicked subcurative PDT and elicited a 1.6- and 2.1-fold increase, respectively, in secreted VEGF 24 hours following PDT. Intracellular VEGF protein measurement and VEGF mRNA showed a 1.4- and 1.6-fold increase only at 0.5 J/cm(2). In vivo subcurative PDT showed an increase in VEGF by both immunohistochemistry and ELISA. In vitro analysis showed no activation of hypoxia-inducible factor-1alpha (HIF-1alpha) or cyclooxygenase-2 (COX-2) following subcurative PDT; furthermore, small interfering RNA inhibition of HIF-1alpha and COX-2 inhibitor treatment had no effect on PDT induction of VEGF. PDT in the presence of phosphatidylinositol 3-kinase/AKT inhibitor or mitogen-activated protein kinase (MAPK)/extracellular signal-regulated kinase inhibitor still induced VEGF. However, subcurative PDT increased phosphorylated p38 and stress-activated protein kinase/c-Jun NH(2)-terminal kinase. The p38 MAPK inhibitor abolished PDT induction of VEGF. The results establish the importance of VEGF in subcurative BPD-PDT of prostate cancer and suggest possible molecular pathways for its induction. These findings should provide the basis for the development of molecular-based interventions for enhancing PDT and merit further studies.


Subject(s)
Photochemotherapy/methods , Prostatic Neoplasms/drug therapy , Prostatic Neoplasms/metabolism , Vascular Endothelial Growth Factor A/biosynthesis , Animals , Cell Line, Tumor , Cyclooxygenase 2/metabolism , Cyclooxygenase 2 Inhibitors/pharmacology , Humans , Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , MAP Kinase Signaling System/drug effects , Male , Mice , Mice, SCID , Photosensitizing Agents/pharmacology , Porphyrins/pharmacology , Prostatic Neoplasms/genetics , RNA, Messenger/biosynthesis , RNA, Messenger/genetics , Vascular Endothelial Growth Factor A/genetics , Vascular Endothelial Growth Factor A/metabolism , Xenograft Model Antitumor Assays
20.
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
SELECTION OF CITATIONS
SEARCH DETAIL
...