Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 16 de 16
Filter
Add more filters










Publication year range
1.
ACS Biomater Sci Eng ; 10(6): 3825-3832, 2024 06 10.
Article in English | MEDLINE | ID: mdl-38722049

ABSTRACT

In recent years, a novel treatment method for cancer has emerged, which is based on the starvation of tumors of amino acids like arginine. The deprivation of arginine in serum is based on enzymatic degradation and can be realized by arginine deaminases like the l-amino acid oxidase found in the ink toxin of the sea hare Aplysia punctata. Previously isolated from the ink, the l-amino acid oxidase was described to oxidate the essential amino acids l-lysine and l-arginine to their corresponding deaminated alpha-keto acids. Here, we present the recombinant production and functionalization of the amino acid oxidase Aplysia punctata ink toxin (APIT). PEGylated APIT (APIT-PEG) increased the blood circulation time. APIT-PEG treatment of patient-derived xenografted mice shows a significant dose-dependent reduction of tumor growth over time mediated by amino acid starvation of the tumor. Treatment of mice with APIT-PEG, which led to deprivation of arginine, was well tolerated.


Subject(s)
Aplysia , Arginine , Lysine , Polyethylene Glycols , Animals , Arginine/pharmacology , Arginine/chemistry , Lysine/pharmacology , Lysine/chemistry , Polyethylene Glycols/chemistry , Polyethylene Glycols/pharmacology , Humans , Mice , Xenograft Model Antitumor Assays , Marine Toxins/pharmacology , Marine Toxins/therapeutic use , Marine Toxins/chemistry , Recombinant Proteins/pharmacology , Recombinant Proteins/therapeutic use , L-Amino Acid Oxidase/pharmacology , L-Amino Acid Oxidase/metabolism , L-Amino Acid Oxidase/chemistry , Female , Cell Line, Tumor
2.
Horm Res Paediatr ; 94(7-8): 239-250, 2021.
Article in English | MEDLINE | ID: mdl-34438400

ABSTRACT

BACKGROUND/AIMS: Diagnosis of growth hormone deficiency (GHD) in children requires the use of provocative growth hormone (GH) stimulation tests, which can have limited reliability and are potentially contraindicated in some patients. This is the first paediatric study to test the safety, tolerability, and pharmacokinetics (PK)/pharmacodynamics (PD) of macimorelin, an oral GH secretagogue, approved for diagnosis of adult GHD. METHODS: In this open-label, group comparison, single-dose escalation trial (EudraCT 2018-001988-23), sequential cohorts of patients (C1-C3) received ascending single doses of macimorelin: 0.25 (C1), 0.5 (C2), and 1.0 (C3) mg/kg. Primary endpoints were safety and tolerability, and secondary endpoints were PK/PD. RESULTS: Twenty-four patients aged between 2 and <18 with suspected GHD participated in the study. No macimorelin-related adverse events were reported, and macimorelin was well tolerated. Plasma macimorelin concentrations increased with dose: mean areas under the curve were 6.69 (C1), 18.02 (C2), and 30.92 (C3) h × ng/mL; mean maximum concentrations were 3.46 (C1), 8.13 (C2), and 12.87 (C3) ng/mL. GH concentration increased following macimorelin administration: mean times of maximum measured concentration were 52.5 (C1), 37.5 (C2), and 37.5 (C3) min. CONCLUSION: All 3 doses of macimorelin had excellent safety and tolerability with PK/PD profiles in expected ranges. These results support the use of 1.0 mg/mL macimorelin in a Phase 3 test validation trial in children.


Subject(s)
Dose-Response Relationship, Drug , Growth Hormone , Indoles/administration & dosage , Pediatrics , Tryptophan/analogs & derivatives , Child , Female , Ghrelin , Growth Hormone/deficiency , Growth Hormone/drug effects , Humans , Indoles/pharmacokinetics , Male , Reproducibility of Results , Surveys and Questionnaires , Tryptophan/administration & dosage , Tryptophan/pharmacokinetics
3.
Cancer Chemother Pharmacol ; 81(2): 291-304, 2018 02.
Article in English | MEDLINE | ID: mdl-29204687

ABSTRACT

PURPOSE: Zoptarelin doxorubicin is a fusion molecule of the chemotherapeutic doxorubicin and a luteinizing hormone-releasing hormone receptor (LHRHR) agonist, designed for drug targeting to LHRHR positive tumors. The aim of this study was to establish a physiologically based pharmacokinetic (PBPK) parent-metabolite model of zoptarelin doxorubicin and to apply it for drug-drug interaction (DDI) potential analysis. METHODS: The PBPK model was built in a two-step procedure. First, a model for doxorubicin was developed, using clinical data of a doxorubicin study arm. Second, a parent-metabolite model for zoptarelin doxorubicin was built, using clinical data of three different zoptarelin doxorubicin studies with a dosing range of 10-267 mg/m2, integrating the established doxorubicin model. DDI parameters determined in vitro were implemented to predict the impact of zoptarelin doxorubicin on possible victim drugs. RESULTS: In vitro, zoptarelin doxorubicin inhibits the drug transporters organic anion-transporting polypeptide 1B3 (OATP1B3) and organic cation transporter 2 (OCT2). The model was applied to evaluate the in vivo inhibition of these transporters in a generic manner, predicting worst-case scenario decreases of 0.5% for OATP1B3 and of 2.5% for OCT2 transport rates. Specific DDI simulations using PBPK models of simvastatin (OATP1B3 substrate) and metformin (OCT2 substrate) predict no significant changes of the plasma concentrations of these two victim drugs during co-administration. CONCLUSIONS: The first whole-body PBPK model of zoptarelin doxorubicin and its active metabolite doxorubicin has been successfully established. Zoptarelin doxorubicin shows no potential for DDIs via OATP1B3 and OCT2.


Subject(s)
Antineoplastic Agents/pharmacokinetics , Doxorubicin/analogs & derivatives , Gonadotropin-Releasing Hormone/analogs & derivatives , Adult , Aged , Aged, 80 and over , Antineoplastic Agents/adverse effects , Biotransformation , Computer Simulation , Doxorubicin/adverse effects , Doxorubicin/pharmacokinetics , Drug Interactions , Female , Gonadotropin-Releasing Hormone/adverse effects , Gonadotropin-Releasing Hormone/pharmacokinetics , Humans , Hydroxymethylglutaryl-CoA Reductase Inhibitors/pharmacokinetics , Hypoglycemic Agents/pharmacokinetics , Male , Metformin/pharmacokinetics , Middle Aged , Models, Biological , Octamer Transcription Factor-2 , Simvastatin/pharmacokinetics , Solute Carrier Organic Anion Transporter Family Member 1B3/metabolism
4.
Bioorg Med Chem Lett ; 26(10): 2408-2412, 2016 05 15.
Article in English | MEDLINE | ID: mdl-27072910

ABSTRACT

Introducing a second chiral center on our previously described 1,2,4-triazole, allowed us to increase diversity and elongate the 'C-terminal part' of the molecule. Therefore, we were able to explore mimics of the substance P analogs described as inverse agonists. Some compounds presented affinities in the nanomolar range and potent biological activities, while one exhibited a partial inverse agonist behavior similar to a Substance P analog.


Subject(s)
Receptors, Ghrelin/metabolism , Triazoles/chemistry , Fluorescence Resonance Energy Transfer , Indoles/chemistry , Indoles/pharmacology , Inhibitory Concentration 50 , Ligands , Receptors, Ghrelin/agonists , Structure-Activity Relationship , Substance P/chemistry , Tryptophan/analogs & derivatives , Tryptophan/chemistry , Tryptophan/pharmacology
5.
Bioorg Med Chem Lett ; 25(1): 20-4, 2015 Jan 01.
Article in English | MEDLINE | ID: mdl-25435152

ABSTRACT

Ghrelin receptor ligands based on a trisubstituted 1,2,4-triazole scaffold were recently synthesized and evaluated for their in vitro affinity for the GHS-R1a receptor and their biological activity. In this study, replacement of the α-aminoisobutyryl (Aib) moiety (a common feature present in numerous growth hormone secretagogues described in the literature) by aromatic and heteroaromatic groups was explored. We found potent antagonists incorporating the picolinic moiety in place of the Aib moiety. In an attempt to increase affinity and activity of our lead compound 2, we explored the modulation of the pyridine ring. Herein we report the design and the structure-activity relationships study of these new ghrelin receptor ligands.


Subject(s)
Receptors, Ghrelin/antagonists & inhibitors , Receptors, Ghrelin/metabolism , Triazoles/chemical synthesis , Triazoles/metabolism , Animals , Cell Line , Humans , Mice , Protein Binding/physiology , Structure-Activity Relationship , Triazoles/pharmacology
6.
Bioorg Med Chem Lett ; 24(16): 3748-52, 2014 Aug 15.
Article in English | MEDLINE | ID: mdl-25022204

ABSTRACT

A novel series of (7-aryl-1,5-naphthyridin-2-yl)ureas was discovered as dual ERK2 and Aurora B kinases inhibitors. Several analogues were active at micromolar and submicromolar range against ERK2 and Aurora B, associated with very promising antiproliferative activity toward various cancer cell lines. Synthesis, structure activity relationship and docking study are reported. In vitro ADME properties and safety data are also discussed.


Subject(s)
Antineoplastic Agents/pharmacology , Aurora Kinase B/antagonists & inhibitors , Drug Discovery , Mitogen-Activated Protein Kinase 1/antagonists & inhibitors , Protein Kinase Inhibitors/pharmacology , Urea/pharmacology , Antineoplastic Agents/chemical synthesis , Antineoplastic Agents/chemistry , Aurora Kinase B/metabolism , Cell Line, Tumor , Cell Proliferation/drug effects , Dose-Response Relationship, Drug , Drug Screening Assays, Antitumor , HCT116 Cells , Humans , Mitogen-Activated Protein Kinase 1/metabolism , Models, Molecular , Molecular Structure , Protein Kinase Inhibitors/chemical synthesis , Protein Kinase Inhibitors/chemistry , Structure-Activity Relationship , Urea/analogs & derivatives , Urea/chemistry
7.
Anticancer Agents Med Chem ; 14(4): 629-35, 2014 May.
Article in English | MEDLINE | ID: mdl-24628236

ABSTRACT

Perifosine treatment exhibits a complex molecular response including the inhibition of Akt or the induction of apoptosis via clustering of death receptors in lipid rafts. However, the molecular response can vary between different tumor entities and the contribution of each target pathway to the activity of Perifosine might be distinct depending on the tumor entity or the agent combined with Perifosine. In this review we discuss the current view on the mechanism of action of perifosine in cancer and the contribution of the molecular targets of Perifosine to its activity.


Subject(s)
Antineoplastic Agents/pharmacology , Antineoplastic Agents/therapeutic use , Neoplasms/drug therapy , Phosphorylcholine/analogs & derivatives , Animals , Antineoplastic Agents/chemistry , Antineoplastic Agents/metabolism , Apoptosis/drug effects , Cell Line, Tumor , Cell Membrane/drug effects , Cell Membrane/metabolism , Humans , Lipid Metabolism/drug effects , Membrane Microdomains/drug effects , Membrane Microdomains/metabolism , Neoplasms/metabolism , Neoplasms/pathology , Neoplastic Stem Cells/drug effects , Neoplastic Stem Cells/metabolism , Neoplastic Stem Cells/pathology , Phosphorylcholine/chemistry , Phosphorylcholine/metabolism , Phosphorylcholine/pharmacology , Phosphorylcholine/therapeutic use , Proto-Oncogene Proteins c-akt/metabolism , Receptors, Death Domain/metabolism , Signal Transduction
8.
ChemMedChem ; 9(1): 217-32, 2014 Jan.
Article in English | MEDLINE | ID: mdl-24273104

ABSTRACT

As part of our research projects to identify new chemical entities of biological interest, we developed a synthetic approach and the biological evaluation of (7-aryl-1,5-naphthyridin-4-yl)ureas as a novel class of Aurora kinase inhibitors for the treatment of malignant diseases based on pathological cell proliferation. 1,5-Naphthyridine derivatives showed excellent inhibitory activities toward Aurora kinases A and B, and the most active compound, 1-cyclopropyl-3-[7-(1-methyl-1H-pyrazol-4-yl)-1,5-naphthyridin-4-yl]urea (49), displayed IC50 values of 13 and 107 nM against Aurora kinases A and B, respectively. In addition, the selectivity toward a panel of seven cancer-related protein kinases was highlighted. In vitro ADME properties were also determined in order to rationalize the difficulties in correlating antiproliferative activity with Aurora kinase inhibition. Finally, the good safety profile of these compounds imparts promising potential for their further development as anticancer agents.


Subject(s)
Aurora Kinase A/antagonists & inhibitors , Aurora Kinase B/antagonists & inhibitors , Protein Kinase Inhibitors/analogs & derivatives , Urea/analogs & derivatives , Animals , Aurora Kinase A/genetics , Aurora Kinase A/metabolism , Aurora Kinase B/genetics , Aurora Kinase B/metabolism , Caco-2 Cells , Cell Line, Tumor , Cell Proliferation/drug effects , Drug Evaluation, Preclinical , HCT116 Cells , Half-Life , Humans , Mice , Microsomes, Liver/metabolism , Naphthyridines/chemistry , Protein Binding , Protein Kinase Inhibitors/pharmacokinetics , Protein Kinase Inhibitors/pharmacology , Recombinant Proteins/biosynthesis , Recombinant Proteins/chemistry , Recombinant Proteins/genetics , Structure-Activity Relationship , Urea/pharmacokinetics , Urea/pharmacology
9.
Oncol Rep ; 22(2): 321-6, 2009 Aug.
Article in English | MEDLINE | ID: mdl-19578772

ABSTRACT

Neovascular targeting is an established approach for the therapy of prostate cancer (PCa). Cationic liposomes have been shown to be absorbed by immature vascular endothelial cells due to negative electric charge of their outer cell membrane. We aimed to evaluate the antitumoural efficacy of paclitaxel encapsulated in cationic liposomes for the treatment of PCa. Tumours were generated by subcutaneous injection of 10(6) MatLu tumour cells into the right hind leg of 21 male Copenhagen rats. After tumour growth, the animals were treated by an i.v. infusion with either 5% glucose (Gl), paclitaxel (Pax), cationic liposomes (CL) or paclitaxel encapsulated in cationic liposomes (EndoTAG-1) on days 12, 14, 16 and 19. Treatment was initiated on day 12 after tumour inoculation at mean tumour volumes of 0.31+/-0.13 mm(3). On the last day of treatment, animals treated with EndoTAG-1 had the significantly lowest tumour volumes with 2.49+/-0.84 cm(3) vs. Pax (5.59+/-0.45 cm(3)) vs. CL (3.87+/-1.25 cm(3)) vs. GL (5.17+/-1.70 cm(3)). The quantification of MVD showed the lowest count for EndoTAG-1-treated tumours (11.78+/-2.68 vessels/mm(2)) followed by Gl (15.64+/-6.68 vessels/mm(3)), Pax (18.22+/-9.50 vessels/mm(3)) and CL (40.9+/-32.8 vessels/mm(3)). The data confirm that neovascular targeting with EndoTAG-1 is a promising new method for the treatment of PCa by reducing the primary tumour mass and demonstrating benefits in the suppression of angiogenesis in comparison with the conventional treatment.


Subject(s)
Antineoplastic Agents, Phytogenic/administration & dosage , Paclitaxel/administration & dosage , Prostatic Neoplasms/drug therapy , Animals , Cell Line, Tumor , Liposomes , Male , Prostatic Neoplasms/blood supply , Prostatic Neoplasms/pathology , Rats
10.
Clin Cancer Res ; 14(14): 4603-11, 2008 Jul 15.
Article in English | MEDLINE | ID: mdl-18628475

ABSTRACT

PURPOSE: Paclitaxel encapsulated in cationic liposomes (EndoTAG-1) is a vascular targeting formulation for the treatment of solid tumors. It triggers intratumoral microthrombosis, causing significant inhibition of tumor perfusion and tumor growth associated with endothelial cell apoptosis. Here, we quantified the effects of repeated EndoTAG-1 therapy on tumor microvascular leakiness with respect to leukocyte-endothelial cell interactions, the targeting property of cationic liposomes, and the therapeutic combination with conventional cisplatin chemotherapy. EXPERIMENTAL DESIGN: Using dorsal skinfold chamber preparations in Syrian Golden hamsters, in vivo fluorescence microscopy experiments were done after repeated EndoTAG-1 treatment of A-Mel-3 tumors. Controls received glucose, paclitaxel alone, or cationic liposomes devoid of paclitaxel. Extravasation of rhodamine-labeled albumin was measured to calculate microvessel permeability, and intratumoral leukocyte-endothelial cell interactions were quantified. Subcutaneous tumor growth was evaluated after combination therapy followed by histologic analysis. RESULTS: Microvascular permeability was significantly increased only after treatment with EndoTAG-1, whereas intratumoral leukocyte-endothelial cell interactions were not affected by any treatment. In separate skinfold chamber experiments, fluorescently labeled cationic liposomes kept their targeting property for tumor endothelial cells after repeated EndoTAG-1 treatment and no signs of extravasation were observed. Subcutaneous A-Mel-3 tumor growth was significantly inhibited by the combination of cisplatin and EndoTAG-1. CONCLUSIONS: These data show that vascular targeting with EndoTAG-1 increases tumor microvessel leakiness probably due to vascular damage. This mechanism is not mediated by inflammatory leukocyte-endothelial cell interactions. Manipulating the blood-tumor barrier by repeated tumor microvessel targeting using EndoTAG-1 can effectively be combined with tumor cell-directed conventional cisplatin chemotherapy.


Subject(s)
Antineoplastic Agents, Phytogenic/administration & dosage , Capillaries/drug effects , Capillary Permeability/drug effects , Melanoma, Experimental/drug therapy , Neovascularization, Pathologic/drug therapy , Paclitaxel/administration & dosage , Animals , Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Cations , Cisplatin/administration & dosage , Cricetinae , Liposomes , Male , Melanoma, Experimental/blood supply , Mesocricetus
11.
Int J Cancer ; 122(2): 452-60, 2008 Jan 15.
Article in English | MEDLINE | ID: mdl-17918179

ABSTRACT

Paclitaxel encapsulated in cationic liposomes (EndoTAG-1) significantly impairs tumor growth by a significant reduction of functional tumor microcirculation and induction of endothelial cell apoptosis. The aim of the study was to analyze whether platelet activation within the tumor microcirculation contributes to the antivascular effects of vascular targeting chemotherapy using EndoTAG-1. In vitro, FACS analysis revealed a significant activation of platelets upon treatment with EndoTAG-1. In vivo, using A-Mel-3 tumors in Syrian Golden hamsters equipped with dorsal skinfold chamber preparations, the contribution of platelets to the antivascular effects of EndoTAG-1 was evaluated by fluorescence and laser-scanning microscopy. Immediately after a single treatment with EndoTAG-1 or cationic liposomes devoid of paclitaxel, an increase of platelet adherence in tumor microvessels was observed. This was accompanied by an acute impairment of the microcirculation within the treated tumors leading to reduced tumor perfusion. After repetitive therapy, an increase of platelet adherence and subsequent tumor microvessel occlusions occurred only after treatment with EndoTAG-1. Comparing to "tumor free" normal tissue controls these microthromboses were tumor selective. Significantly disbalancing the coagulation system within tumors by targeted induction of microthromboses within the tumor microcirculation appears to be an important mechanism of EndoTAG-1 therapy.


Subject(s)
Antineoplastic Agents/pharmacology , Blood Platelets/metabolism , Drug Delivery Systems , Liposomes/chemistry , Paclitaxel/administration & dosage , Animals , Apoptosis , Cricetinae , Endothelial Cells/pathology , Flow Cytometry , Mesocricetus , Microcirculation , Microscopy, Confocal , Microscopy, Fluorescence , Paclitaxel/pharmacology , Platelet Adhesiveness
12.
Biol Chem ; 386(2): 167-75, 2005 Feb.
Article in English | MEDLINE | ID: mdl-15843161

ABSTRACT

Microvascular endothelial cells from human neonatal foreskin were grown in vitro until a three-dimensional network of capillary-like structures was formed. All stages of the angiogenic cascade could be observed in this in vitro model, including the formation of an internal lumen. The microscopy focused on morphology, formation of an internal lumen, role of the extracellular matrix, polarity of the cells, and the time-course of the angiogenic cascade. Bright-field microscopy revealed cells arranged circularly side by side and the internal lumen of capillary-like structures was verified by electron microscopy. Immunolabeling revealed a peritubular localization of collagen IV. Reporter gene expression after the formation of capillary-like structures was marginally higher than control expression, but clearly lower than the expression of cells at the stage of proliferation. Highest transfection efficiencies were obtained using vectors with the CMV promoter and the long fragment of the Ets-1 promoter. This is a first study of transfection efficiencies mapped for stages of in vitro angiogenesis. We describe here the morphological features of a long-term in vitro model of angiogenesis of human microvascular endothelial cells that could be used for transfection studies, without the provision of an extracellular matrix substrate. The cells self-create their own extracellular matrix to proliferate and form a three-dimensional network of capillary-like structures with an internal lumen.


Subject(s)
Endothelial Cells/cytology , Endothelium, Vascular/cytology , Neovascularization, Physiologic , Transfection , Capillaries/growth & development , Capillaries/ultrastructure , Cell Polarity , Collagen Type IV/biosynthesis , Extracellular Matrix/ultrastructure , Humans , Infant, Newborn , Luciferases/biosynthesis , Male , Microscopy , Models, Biological
13.
Int J Cancer ; 110(1): 117-24, 2004 May 20.
Article in English | MEDLINE | ID: mdl-15054876

ABSTRACT

Cationic liposomes have been shown to be internalized selectively by angiogenic tumor endothelial cells after intravenous injection. Therefore, encapsulation of cytotoxic substances in cationic liposomes is a new approach to target tumor vasculature. It was the aim of our study to quantify the effects of paclitaxel encapsulated in cationic liposomes (MBT-0206) on tumor microvasculature and growth in vivo. Experiments were performed in the dorsal skinfold chamber preparation of Syrian Golden hamsters bearing syngeneic A-Mel-3 melanomas. Tumors were treated with intravenous infusion of MBT-0206 (20 mM) resulting in an effective paclitaxel dose of 5 mg/kg body weight (b.w.). Control animals received conventional paclitaxel in Cremophor EL (Taxol(R); 5 mg/kg b.w.), unloaded cationic liposomes (20 mM) or the solvent 5% glucose, respectively. Using intravital microscopy, tumor growth and effects on intratumoral microvasculature were analyzed. Tumor growth was significantly retarded after treatment with MBT-0206 compared to the treatment with paclitaxel. Analysis of intratumoral microcirculation revealed a reduced functional vessel density in tumors after application of liposomal paclitaxel. At the end of the observation time, vessel diameters were significantly smaller in animals treated with paclitaxel encapsulated in cationic liposomes while red blood cell velocity was less affected. This resulted in a significantly reduced blood flow in vessel segments and a reduced microcirculatory perfusion index in these animals. Histochemical TUNEL stain was vessel-associated after treatment with liposomal paclitaxel in contrast to few apoptotic tumor cells in the control groups. Our data demonstrate that encapsulation of paclitaxel in cationic liposomes significantly increased the antitumoral efficacy of the drug. Remarkable microcirculatory changes indicate that encapsulation of paclitaxel in cationic liposomes resulted in a mechanistic switch from tumor cell toxicity to an antivascular therapy.


Subject(s)
Melanoma, Experimental/blood supply , Melanoma, Experimental/drug therapy , Neovascularization, Pathologic/drug therapy , Paclitaxel/administration & dosage , Animals , Cricetinae , Drug Carriers , Erythrocytes/drug effects , In Situ Nick-End Labeling , Liposomes , Male , Melanoma, Experimental/pathology , Mesocricetus , Microcirculation/drug effects , Regional Blood Flow/drug effects
14.
Clin Cancer Res ; 9(6): 2335-41, 2003 Jun.
Article in English | MEDLINE | ID: mdl-12796403

ABSTRACT

PURPOSE: Cationic liposomes have been shown to selectively target tumor endothelial cells. Therefore, the encapsulation of antineoplastic drugs into cationic liposomes is a promising tool to improve selective drug delivery by targeting tumor vasculature. It was the aim of our study to evaluate tumor selectivity and antitumoral efficacy of paclitaxel encapsulated in cationic liposomes in comparison with the free drug paclitaxel (Taxol(R)) in vivo. EXPERIMENTAL DESIGN: Experiments evaluating tumor selectivity were carried out in male Syrian golden hamsters bearing the amelanotic hamster melanoma A-Mel-3 in dorsal skinfold preparations. Growth of tumor cells was observed after s.c. inoculation (day 0). On days 5, 7, 9, 12, 14, and 16, animals were treated by continuous i.v. infusion over 90 min with 5% glucose, Taxol(R), unloaded cationic liposomes, or paclitaxel encapsulated into cationic liposomes (LipoPac), respectively (lipid dose, 150 mg/kg body weight; paclitaxel dose, 5 mg/kg body weight). Tumor volumes and presence of regional lymph node metastases were quantified. RESULTS: Vascular targeting of rhodamine-labeled cationic liposomes was maintained after encapsulation of paclitaxel as revealed by in vivo fluorescence microscopy (ratio of dye concentration, tumor:normal tissue = 3:1). The s.c. tumor growth revealed a remarkable retardation of tumor growth after treatment with LipoPac (1.7 +/- 0.3 cm(3)). In contrast, control tumors showed exponential tumor growth [tumor volume at the end of the observation period (mean +/- SE): 5% glucose, 17.7 +/- 1.9 cm(3); unloaded cationic liposomes, 10.0 +/- 1.6 cm(3); Taxol(R), 10.7 +/- 1.7 cm(3)]. In addition, the appearance of regional lymph node metastases was significantly delayed by treatment with paclitaxel encapsulated into cationic liposomes in comparison with all other groups. CONCLUSIONS: The data suggest that cationic liposomes are a powerful tool for selective and efficient drug delivery to tumor microvessels. This may serve as proof of the concept of neovascular tumor targeting therapy by cationic liposomes.


Subject(s)
Neoplasms, Experimental/blood supply , Neoplasms, Experimental/drug therapy , Paclitaxel/administration & dosage , Animals , Cricetinae , Liposomes , Lymphatic Metastasis , Male , Mesocricetus , Microscopy, Fluorescence , Paclitaxel/pharmacokinetics , Paclitaxel/pharmacology
15.
Int J Cancer ; 105(4): 561-7, 2003 Jul 01.
Article in English | MEDLINE | ID: mdl-12712451

ABSTRACT

Recently, cationic liposomes have been shown to preferentially target the angiogenic endothelium of tumors. It was the aim of our study to investigate the influence of liposomal surface charge on the uptake and kinetics of liposomes into solid tumors and tumor vasculature. Experiments were performed in the amelanotic hamster melanoma A-Mel-3 growing in the dorsal skinfold chamber preparation of male Syrian golden hamsters. Fluorescently labeled liposomes with different surface charge were prepared. Accumulation of i.v. injected liposomes was assessed by quantitative intravital fluorescence microscopy of tumor and surrounding host tissue. The histological distribution of liposomes was analyzed by double-fluorescence microscopy 20 min after application of fluorescently labeled lectin as a vascular marker. After i.v. application of anionic and neutral liposomes, we observed an almost homogeneous distribution of liposome-induced fluorescence throughout the chamber preparation without specific targeting to tumor tissue. In contrast, cationic liposomes exhibited a significantly enhanced accumulation in tumor tissue and tumor vasculature up to 3-fold compared to surrounding tissue (p<0.05). The histological distribution of neutral and anionic liposomes revealed extravasation 20 min after i.v. injection, while cationic liposomes displayed a highly selective accumulation on the vascular endothelium. In conclusion, cationic liposomes exhibited a preferential uptake in angiogenic tumor vessels and therefore may provide an efficient tool for the selective delivery of diagnostic or therapeutic agents to angiogenic blood vessels of solid tumors. On the other hand, anionic and neutral liposomes may be used as carriers of drugs to the extravascular compartment of tumors due to their extravasation.


Subject(s)
Liposomes/chemistry , Liposomes/pharmacokinetics , Melanoma, Experimental/blood supply , Neovascularization, Pathologic , Animals , Cations , Cricetinae , Diffusion Chambers, Culture , Drug Delivery Systems/methods , Endothelium, Vascular/metabolism , Injections, Intravenous , Liposomes/administration & dosage , Male , Mesocricetus , Surface Properties , Tissue Distribution , Tumor Cells, Cultured
16.
J Invest Dermatol ; 120(3): 476-82, 2003 Mar.
Article in English | MEDLINE | ID: mdl-12603862

ABSTRACT

Paclitaxel is an alkaloid that inhibits endothelial cell proliferation, motility, and tube formation at nanomolar concentrations. Cationic liposome preparations have been shown to target blood vessels. We wished to explore the possibility that paclitaxel encapsulated in cationic liposomes carries paclitaxel to blood vessels and thereby provides an antiangiogenic effect. We used a humanized SCID mouse melanoma model, which allowed us to analyze tumor growth and tumor angiogenesis in an orthotopic tumor model. Here, human melanoma cells grow on human dermis and are in part nourished by human vessels. We show that paclitaxel encapsulated in liposomes prevents melanoma growth and invasiveness and improves survival of mice. Moreover, liposome-encapsulated paclitaxel reduces vessel density at the interface between the tumor and the human dermis and reduces endothelial cell mitosis to background levels. In contrast, equimolar concentrations of paclitaxel solubilized in Cremophor EL(R) had only insignificant effects on tumor growth and did not reduce the mitotic index of endothelium in vivo, although the antiproliferative effect of solubilized paclitaxel in Cremophor EL(R)in vitro was identical to that seen with liposome-coupled paclitaxel. In conclusion, we present a model of how to exploit cytotoxic effects of compounds to prevent tumor growth by using cationic liposomes for targeting an antiproliferative drug to blood vessels.


Subject(s)
Angiogenesis Inhibitors/administration & dosage , Antineoplastic Agents, Phytogenic/administration & dosage , Melanoma, Experimental/blood supply , Melanoma, Experimental/pathology , Neovascularization, Pathologic/drug therapy , Paclitaxel/administration & dosage , Skin Neoplasms/blood supply , Skin Neoplasms/pathology , Animals , Capsules , Cations/analysis , Cell Division/drug effects , Endothelium, Vascular/cytology , Endothelium, Vascular/drug effects , Endothelium, Vascular/pathology , Humans , Liposomes/chemistry , Mice , Mice, SCID , Mitotic Index , Neoplasm Invasiveness/pathology , Tumor Cells, Cultured/pathology
SELECTION OF CITATIONS
SEARCH DETAIL
...