Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 165
Filter
1.
Adv Healthc Mater ; 8(2): e1801160, 2019 01.
Article in English | MEDLINE | ID: mdl-30548428

ABSTRACT

Chronic tympanic membrane (TM) perforations can cause otorrhea. To date, various types of tissue engineering techniques have been applied for the regeneration of chronic TM perforations. However, the application of nanofibers with radially aligned nanostructures and the simultaneous release of growth factors have never been applied in the regeneration of chronic TM perforations. Here, epidermal growth factor (EGF)-releasing radially aligned nanofibrous patches (ERA-NFPs) are developed and applied for the regeneration of chronic perforated TMs. First, radial alignments and the presence of EGF in the ERA-NFPs are analyzed. EGF is confirmed to be released from the ERA-NFPs until 8 weeks. In an in vitro study, cell viability assay, immunocytochemistry, and wound-healing assay indicate rational enhancement of healing by the combination of radial alignments and EGF release. The effect of ERA-NFPs on TM cells is revealed by quantitative real-time polymerase chain reaction. An in vivo animal study shows that the ERA-NFPs effectively stimulates the healing of the chronic TM perforations. The TMs healed by ERA-NFPs show histological properties similar to those of normal TMs. These results indicate that ERA-NFPs may be an efficient platform for the regeneration of chronic TM perforations, laying the foundation for nonsurgical treatments of chronic otitis media.


Subject(s)
Epidermal Growth Factor/pharmacokinetics , Nanofibers/administration & dosage , Nanofibers/chemistry , Tympanic Membrane Perforation/therapy , Animals , Cell Survival/drug effects , Drug Liberation , Epidermal Growth Factor/chemistry , Female , Gene Expression Regulation/drug effects , Guided Tissue Regeneration/methods , Microscopy, Electron, Scanning , Rats, Sprague-Dawley , Spectroscopy, Fourier Transform Infrared
2.
Biointerphases ; 12(1): 010501, 2017 03 21.
Article in English | MEDLINE | ID: mdl-28325051

ABSTRACT

In an effort to rationalize and optimize an antiapoptotic coating combining chondroitin sulfate (CS) and epidermal growth factor (EGF) for vascular applications, the authors here report the comparison of two grafting strategies aiming to display EGF in an oriented fashion on CS. For that purpose, the authors produced, purified, and characterized a chimeric protein corresponding to EGF that was N-terminally fused to a cysteine and a coil peptide. The chimera was covalently immobilized via its free thiol group or captured via coiled-coil interactions at the surface of a biosensor or on a chondroitin sulfate coating in multiwell plates, mimicking the coating that was previously developed by them for stent-graft surfaces. The interactions of grafted EGF with the soluble domain of its receptor or the impact of grafted EGF upon vascular smooth muscle survival in proapoptotic conditions indicated that the coiled-coil based tethering was the best approach to display EGF. These results, combined to direct enzyme-linked immunosorbent assay measurements, indicated that the coiled-coil tethering approach allowed increasing the amount of bioavailable EGF when compared to covalent coupling, rather than the total amount of grafted EGF, while using much lower concentrations of tagged EGF during incubation.


Subject(s)
Chondroitin Sulfates/metabolism , Epidermal Growth Factor/metabolism , Epidermal Growth Factor/pharmacokinetics , Immobilized Proteins/metabolism , Immobilized Proteins/pharmacokinetics , Animals , Biological Availability , Cell Line , Cell Survival/drug effects , Humans , Myocytes, Smooth Muscle/drug effects , Myocytes, Smooth Muscle/physiology , Protein Binding , Rats
3.
J Nanosci Nanotechnol ; 17(4): 2363-369, 2017 Apr.
Article in English | MEDLINE | ID: mdl-29641166

ABSTRACT

Topical administration of growth factors has been suggested as a promising strategy for promoting the healing process and skin regeneration in wound management. However, several restrictions hinder their successful clinical use; specifically, limited percutaneous absorption causes inconsistent efficacy, and various growth factors with specific functionalities are required at different stages of healing. To overcome these shortcomings, previously we have constructed highly skin-permeable analogues of epidermal growth factor (EGF), insulin-like growth factor-I (IGF-I), and platelet-derived growth factor-A (PDGF-A) (LMWP-EGF, LMWP-IGF-I and LMWP-PDGF-A) by genetically conjugating the low-molecular-weight protamine (LMWP) to their N-terminus. In the present study, we determined the optimal concentration ratio of these growth factors by investigating In Vitro cell proliferation and the scratch wound repairing assay. After confirming synergetic effects of growth factors in combinations, we developed a topical delivery system consisting of a nanoemulsion (NE)-dispersed polyvinylpyrrolidone hydrogel loaded with all three growth factors. In Vitro permeability studies were also performed to assess whether the LMWP-conjugated growth factors in the formulation enhanced their skin permeation compared to native growth factors. Combinations of native or LMWP-fused growth factors significantly promoted fibroblast proliferation and scratch wound recovery, and the synergy of LMWP-EGF, LMWP-IGF-I and LMWP-PDGF-A was optimal at a ratio of 100:100:10 by concentration. The growth factor combination-loaded NE appeared to be spherical under cryo-transmission electron microscopy and the average droplet diameter was 127±4.30 nm. The LMWP-conjugated growth factors allowed significantly higher skin permeation than native growth factors from the NE-dispersed hydrogel. Thus, the LMWP-conjugated growth factor combination-loaded NE-dispersed hydrogel is expected to induce more rapid and prolonged wound healing.


Subject(s)
Drug Delivery Systems , Epidermal Growth Factor , Hydrogel, Polyethylene Glycol Dimethacrylate/chemistry , Nanostructures/chemistry , Platelet-Derived Growth Factor , Administration, Topical , Animals , Emulsions/chemistry , Epidermal Growth Factor/administration & dosage , Epidermal Growth Factor/chemistry , Epidermal Growth Factor/pharmacokinetics , Epidermal Growth Factor/pharmacology , Hydrogel, Polyethylene Glycol Dimethacrylate/pharmacology , Mice , Models, Biological , NIH 3T3 Cells , Platelet-Derived Growth Factor/administration & dosage , Platelet-Derived Growth Factor/chemistry , Platelet-Derived Growth Factor/pharmacokinetics , Platelet-Derived Growth Factor/pharmacology , Skin/chemistry , Skin/metabolism , Skin Absorption/drug effects , Wound Healing/drug effects
4.
Int J Pharm ; 512(1): 204-212, 2016 Oct 15.
Article in English | MEDLINE | ID: mdl-27568495

ABSTRACT

Particle preloading method by first loading proteins onto nano- or microparticles and then integrating these particles into electrospun polyester nanofibers has been widely used to encapsulate therapeutic proteins into polyester nanofibers. However, poor method design has resulted in unsatisfactory protein delivery performance. For example, the harsh conditions involved in preloading procedures damage the bioactivities of proteins, the improper integration leads to an uneven distribution of particles in nanofibers or insecure attachment of particles to nanofibers, producing uncontrolled protein release profiles. This study aimed to improve the protein delivery performance of polyester nanofibers by rationally designing a particle preloading method. Positively charged chitosan nanoparticles (CNPs) were used as carriers to adsorb negatively charged proteins in mild conditions and as primary barriers for protein release. The polar CNPs were then homogeneously dispersed in a polar polyester solution and subjected to electrospinning. Microscope observations indicated that CNPs were homogeneously embedded within polyester nanofibers. In vitro release behaviour and cell studies showed that proteins retained their bioactivity and could release from polyester nanofibers in a sustained manner for more than 4 weeks without any initial burst. Epidermal growth factor encapsulated in polyester nanofibers enhanced diabetic wound healing in vivo, demonstrating an application potential in biomedicine. Other properties of the nanofibers, including composition, wettability, cytotoxicity, and cell adhesion and spreading, were examined in detail as well.


Subject(s)
Drug Delivery Systems/methods , Epidermal Growth Factor/administration & dosage , Epidermal Growth Factor/pharmacokinetics , Nanofibers/chemistry , Polyesters/chemistry , Animals , Cell Adhesion/drug effects , Cell Proliferation/drug effects , Cell Survival , Chitosan/chemistry , Drug Liberation , Epidermal Growth Factor/chemistry , Epidermal Growth Factor/pharmacology , Rats , Wound Healing/drug effects
5.
Int J Radiat Biol ; 92(11): 716-723, 2016 11.
Article in English | MEDLINE | ID: mdl-26999580

ABSTRACT

Purpose Radiolabeled antibodies and peptides hold promise for molecular radiotherapy but are often limited by a low payload resulting in inadequate delivery of radioactivity to tumour tissue and, therefore, modest therapeutic effect. We developed a facile synthetic method of radiolabeling indium-111 (111In) to epidermal growth factor (EGF)-gold nanoparticles (111In-EGF-Au NP) with a high payload. Materials and methods EGF-Au NP were prepared via an interaction between gold and the disulphide bonds of EGF and radiolabeled using 111InCl3. Targeting efficiency was investigated by quantitating internalized radioactivity and by confocal imaging following exposure of MDA-MB-468 (1.3 × 106 EGFR/cell) and MCF-7 (104 EGFR/cell) cells to Cy3-EGF-Au NP. Cytotoxicity was evaluated in clonogenic assays. Results The proportion of total administered radioactivity that was internalized by MDA-MB-468 and MCF-7 cells was 15% and 1.3%, respectively (mixing ratio of EGF:Au of 160). This differential uptake in the two cell lines was confirmed using confocal microscopy. 111In-EGF-Au NP were significantly more radiotoxic to MDA-MB-468 than MCF-7 cells with a surviving fraction of 17.1 ± 4.4% versus 89.8 ± 1.4% (p < 0.001) after exposure for 4 h. Conclusions An 111In-labeled EGF-Au nanosystem was developed. It enabled targeted delivery of a high 111In payload specifically to EGFR-positive cancer cells leading to radiotoxicity that can be exploited for molecularly targeted radiotherapy.


Subject(s)
Epidermal Growth Factor/pharmacokinetics , ErbB Receptors/metabolism , Indium Radioisotopes/administration & dosage , Indium Radioisotopes/pharmacokinetics , Metal Nanoparticles/chemistry , Neoplasms, Experimental/metabolism , Neoplasms, Experimental/radiotherapy , Cell Survival/radiation effects , Coated Materials, Biocompatible , Gold/chemistry , Humans , Indium Radioisotopes/chemistry , MCF-7 Cells , Molecular Targeted Therapy/methods , Nanocapsules/chemistry , Neoplasms, Experimental/pathology , Radiopharmaceuticals/administration & dosage , Radiopharmaceuticals/chemistry , Radiopharmaceuticals/pharmacokinetics , Treatment Outcome
6.
J Pharm Sci ; 105(1): 25-30, 2016 Jan.
Article in English | MEDLINE | ID: mdl-26852838

ABSTRACT

Nanotechnology offers a targeted approach to both imaging and treatment of cancer, the leading cause of death worldwide. Previous studies have found that nanoparticles with a wide variety of coatings initiate an immune response leading to sequestration in the liver and spleen. In an effort to find a nanoparticle platform which does not elicit an immune response, we created 43 nm and 44 nm of gold and silver nanoparticles coated with biomolecules normally produced by the body, α-lipoic acid and the epidermal growth factor (EGF), and have used mass spectroscopy to determine their biodistribution in mouse models, 24 h after tail vein injection. Relative to controls, mouse EGF (mEGF)-coated silver and gold nanoprobes are found at background levels in all organs including the liver and spleen. The lack of sequestration of mEGF-coated nanoprobes in the liver and spleen and the corresponding uptake of control nanoprobes at elevated levels in these organs suggest that the former are not recognized by the immune system. Further studies of cytokine and interleukin levels in the blood are required to confirm avoidance of an immune response.


Subject(s)
ErbB Receptors/metabolism , Gold/pharmacokinetics , Metal Nanoparticles/chemistry , Molecular Probes/pharmacokinetics , Nanomedicine/methods , Animals , Epidermal Growth Factor/chemistry , Epidermal Growth Factor/pharmacokinetics , ErbB Receptors/genetics , Gold/chemistry , Injections, Intravenous , Male , Mass Spectrometry , Mice, Inbred C57BL , Microscopy, Electron, Transmission , Molecular Probes/chemistry , Silver/chemistry , Silver/pharmacokinetics , Surface Properties , Thioctic Acid/chemistry , Thioctic Acid/pharmacokinetics , Tissue Distribution
7.
Bioconjug Chem ; 26(12): 2481-96, 2015 Dec 16.
Article in English | MEDLINE | ID: mdl-26525951

ABSTRACT

The design, preparation, as well as structural and functional characterizations of the recombinant fusion protein hVEGF-EGF as a dual-functional agent that may target both EGFR (R: receptor) and angiogenesis are reported. hVEGF-EGF was found to bind to EGFR more strongly than did EGF, and to bind to VEGFR similarly to VEGF. Mass spectrometry measurements showed that the sites of DTPA (diethylenetriaminepentaacetic acid) conjugated hVEGF-EGF (for radiolabeling) were the same as those of its parent hEGF and hVEGF proteins. All DTPA-conjugated proteins retained similar binding capacities to their respective receptors as compared to their respective parent proteins. In vitro cell binding studies using BAEC (a bovine aortic endothelial cell) and MDA-MB-231 (a human breast cancer) cells expressing both EGFR and VEGFR confirmed similar results. Treating BAEC cells with hVEGF-EGF induced remarkable phosphorylation of EGFR, VEGFR, and their downstream targets ERK1/2. Nevertheless, the radiolabeled (111)In-DTPA-hVEGF-EGF showed cytotoxicity against MDA-MB-231 cells. Pharmacokinetic studies using (111)In-DTPA-hVEGF-EGF in BALB/c nude mice showed that appreciable tracer activities were accumulated in liver and spleen. In all, this study demonstrated that the fusion protein hVEGF-EGF maintained the biological specificity toward both EGFR and VEGFR and may be a potential candidate as a dual-targeting moiety in developing anticancer drugs.


Subject(s)
Antineoplastic Agents/administration & dosage , Drug Carriers/chemistry , Epidermal Growth Factor/chemistry , Vascular Endothelial Growth Factor A/chemistry , Animals , Cattle , Cell Line , Cell Line, Tumor , Drug Carriers/metabolism , Drug Carriers/pharmacokinetics , Drug Delivery Systems , Epidermal Growth Factor/metabolism , Epidermal Growth Factor/pharmacokinetics , Female , Humans , Mice , Mice, Inbred BALB C , Mice, Nude , Neoplasms/drug therapy , Neoplasms/metabolism , Pentetic Acid/chemistry , Pentetic Acid/metabolism , Pentetic Acid/pharmacokinetics , Recombinant Fusion Proteins/chemistry , Recombinant Fusion Proteins/metabolism , Recombinant Fusion Proteins/pharmacokinetics , Vascular Endothelial Growth Factor A/metabolism , Vascular Endothelial Growth Factor A/pharmacokinetics
8.
J Pharm Pharmacol ; 67(11): 1519-27, 2015 Nov.
Article in English | MEDLINE | ID: mdl-26255780

ABSTRACT

OBJECTIVES: This study was conducted to examine the tissue distribution of human recombinant epidermal growth factor (EGF) after multiple intravenous and subcutaneous injections in mice. METHODS: Male BALB/c mice were divided into (1) EGF 1 mg/kg intravenous dose, (2) EGF 5 mg/kg intravenous dose, (3) drug-free intravenous control, (4) EGF 1 mg/kg subcutaneous dose, (5) EGF 5 mg/kg subcutaneous dose and (6) drug-free subcutaneous control groups. EGF and drug-free dosing solutions were injected by intravenous and subcutaneous injections once a day for 3 days. EGF concentrations in serum and tissues of kidney, liver, lung, small intestine and tongue were determined by ELISA. KEY FINDINGS: As the intravenous and subcutaneous doses were increased from 1 to 5 mg/kg, serum Cmax and area under the concentration-time curve (AUC) values were increased dose-proportionally. In lung, tongue and small intestine, increases in AUC were dose-proportional after intravenous injections, but greater than dose-proportional after subcutaneous injections. The fold-increases in Cmax and AUC values were lowest in liver and highest in kidney. CONCLUSION: Based on Cmax and AUC data, the systemic exposure achieved by subcutaneous injections was comparable with that achieved by intravenous injections.


Subject(s)
Epidermal Growth Factor/pharmacokinetics , Recombinant Proteins/pharmacokinetics , Animals , Area Under Curve , Dose-Response Relationship, Drug , Epidermal Growth Factor/administration & dosage , Humans , Injections, Intravenous , Injections, Subcutaneous , Male , Mice , Mice, Inbred BALB C , Recombinant Proteins/administration & dosage , Tissue Distribution
9.
J Biomed Mater Res A ; 103(10): 3374-85, 2015 Oct.
Article in English | MEDLINE | ID: mdl-25856734

ABSTRACT

Controlled delivery of multiple therapeutic agents can be considered as an effective approach in skin tissue engineering. In this study, recombinant human epidermal growth factor (rhEGF) and recombinant human basic fibroblast growth factor (rhbFGF) encapsulated in PLGA microspheres were loaded in hybrid scaffolds of PLGA and PEO. The scaffolds with various formulations were fabricated through electrospinning in order to maintain dual, individual or different release rate of rhEGF and rhbFGF. Morphological, physical and mechanical properties of the scaffold were investigated. The scaffold possessed uniform morphology with an average diameter of 280 nm for PLGA and 760 nm for PEO nanofibers. Furthermore, the mechanical properties of the scaffolds were shown to be akin to those of human skin. Bioactivity of the scaffolds for human skin fibroblasts was evaluated. The HSF acquired significant proliferation and well-spread morphology on the scaffolds particularly in the case of different release rate of rhEGF and rhbFGF which implies the synergistic effect of the growth factors. Additionally, collagen and elastin gene expression was significantly up-regulated in the HSF seeded on the scaffolds in the case of individual delivery of rhEGF and dual delivery of rhEGF and rhbFGF. In conclusion, the prepared scaffolds as a suitable supportive substrate and multiple growth factor delivery system can find extensive utilization in skin tissue engineering.


Subject(s)
Epidermal Growth Factor , Fibroblast Growth Factor 2 , Fibroblasts/metabolism , Regeneration/drug effects , Skin Physiological Phenomena/drug effects , Tissue Scaffolds/chemistry , Cell Line , Delayed-Action Preparations/chemistry , Delayed-Action Preparations/pharmacokinetics , Delayed-Action Preparations/pharmacology , Epidermal Growth Factor/chemistry , Epidermal Growth Factor/pharmacokinetics , Epidermal Growth Factor/pharmacology , Fibroblast Growth Factor 2/chemistry , Fibroblast Growth Factor 2/pharmacokinetics , Fibroblast Growth Factor 2/pharmacology , Humans , Recombinant Proteins/chemistry , Recombinant Proteins/pharmacokinetics , Recombinant Proteins/pharmacology
10.
Int J Nanomedicine ; 9: 1665-76, 2014.
Article in English | MEDLINE | ID: mdl-24729702

ABSTRACT

In this work, an ultrasonic spray freeze-drying (USFD) technique was used to prepare a stable liposomal dry powder for transdermal delivery of recombinant human epithelial growth factor (rhEGF). Morphology, particle size, entrapment efficiency, in vitro release, and skin permeability were systematically compared between rhEGF liposomal dry powder prepared using USFD and that prepared using a conventional lyophilization process. Porous and spherical particles with high specific area were produced under USFD conditions. USFD effectively avoided formation of ice crystals, disruption of the bilayer structure, and drug leakage during the liposome drying process, and maintained the stability of the rhEGF liposomal formulation during storage. The reconstituted rhEGF liposomes prepared from USFD powder did not show significant changes in morphology, particle size, entrapment efficiency, or in vitro release characteristics compared with those of rhEGF liposomes before drying. Moreover, the rhEGF liposomal powder prepared with USFD exhibited excellent enhanced penetration in ex vivo mouse skin compared with that for powder prepared via conventional lyophilization. The results suggest that ultrasonic USFD is a promising technique for the production of stable protein-loaded liposomal dry powder for application to the skin.


Subject(s)
Epidermal Growth Factor/administration & dosage , Epidermal Growth Factor/pharmacokinetics , Freeze Drying/instrumentation , Liposomes/chemical synthesis , Liposomes/pharmacokinetics , Skin Absorption/physiology , Sonication/instrumentation , Administration, Cutaneous , Animals , Desiccation/instrumentation , Desiccation/methods , Diffusion , Epidermal Growth Factor/chemistry , Equipment Design , Equipment Failure Analysis , Freeze Drying/methods , Gases/chemistry , Gases/radiation effects , High-Energy Shock Waves , Humans , Liposomes/administration & dosage , Mice , Mice, Inbred BALB C , Mice, Nude , Particle Size , Powders , Recombinant Proteins/administration & dosage , Recombinant Proteins/chemistry , Recombinant Proteins/pharmacokinetics , Sonication/methods
11.
Int J Nanomedicine ; 9: 273-87, 2014.
Article in English | MEDLINE | ID: mdl-24421639

ABSTRACT

Superparamagnetic iron oxide nanoparticles (SPIONs) conjugated with recombinant human epidermal growth factor (SPION-EGF) were studied as a potential agent for magnetic resonance imaging contrast enhancement of malignant brain tumors. Synthesized conjugates were characterized by transmission electron microscopy, dynamic light scattering, and nuclear magnetic resonance relaxometry. The interaction of SPION-EGF conjugates with cells was analyzed in a C6 glioma cell culture. The distribution of the nanoparticles and their accumulation in tumors were assessed by magnetic resonance imaging in an orthotopic model of C6 gliomas. SPION-EGF nanosuspensions had the properties of a negative contrast agent with high coefficients of relaxation efficiency. In vitro studies of SPION-EGF nanoparticles showed high intracellular incorporation and the absence of a toxic influence on C6 cell viability and proliferation. Intravenous administration of SPION-EGF conjugates in animals provided receptor-mediated targeted delivery across the blood-brain barrier and tumor retention of the nanoparticles; this was more efficient than with unconjugated SPIONs. The accumulation of conjugates in the glioma was revealed as hypotensive zones on T2-weighted images with a twofold reduction in T2 relaxation time in comparison to unconjugated SPIONs (P<0.001). SPION-EGF conjugates provide targeted delivery and efficient magnetic resonance contrast enhancement of EGFR-overexpressing C6 gliomas.


Subject(s)
Brain Neoplasms/drug therapy , Brain Neoplasms/metabolism , Dextrans/administration & dosage , Dextrans/chemistry , Epidermal Growth Factor/pharmacokinetics , Glioma/drug therapy , Glioma/metabolism , Magnetite Nanoparticles/administration & dosage , Magnetite Nanoparticles/chemistry , Animals , Apoptosis , Brain Neoplasms/pathology , Cell Line, Tumor , Cell Survival , Dextrans/ultrastructure , Epidermal Growth Factor/chemistry , Epidermal Growth Factor/genetics , Glioma/pathology , Magnetite Nanoparticles/ultrastructure , Nanocapsules/administration & dosage , Nanocapsules/chemistry , Nanocapsules/ultrastructure , Rats , Recombinant Proteins/chemistry , Recombinant Proteins/pharmacokinetics , Treatment Outcome
12.
Protein Pept Lett ; 21(6): 550-5, 2014 Jun.
Article in English | MEDLINE | ID: mdl-24364869

ABSTRACT

TD1, a peptide chaperone consisting of the sequence ACSSSPHKHCG, has been shown to facilitate transdermal delivery for protein molecules via either co-administration or the fusion approach. We previously reported that a single TD1 motif, fused to the N-terminus of human epidermal growth factor (hEGF) can significantly enhance the transdermal efficiency of the recombinant EGF protein. In an effort to further increase the transdermal efficiency, we have created EGF fusion proteins harboring dual TD1 motifs: TD1-hEGF-TD1, containing one TD1 motif at both the N- and the Cterminus, and TD1-TD1-hEGF, containing two tandem TD1 motifs at the N-terminus. Both TD1-hEGF-TD1 and TD1- TD1-hEGF proteins, expressed in Escherichia coli and purified to apparent homogeneity, exhibited biological activity similar to unmodified hEGF, as revealed by their relative abilities to stimulate fibroblast growth, promote fibroblast migration, and activate the MAP kinase signaling cascade. On the other hand, both TD1-hEGF-TD1 and TD1-TD1-hEGF proteins exhibited a transdermal efficiency enhancement. The improvement was >5-fold compared to unmodified hEGF and 3-fold over the hEGF fusion protein with only one TD1 motif attached. These findings provided proof-of-concept for improving transdermal delivery of protein actives through rational protein design.


Subject(s)
Epidermal Growth Factor/administration & dosage , Epidermal Growth Factor/pharmacokinetics , Recombinant Fusion Proteins/administration & dosage , Recombinant Fusion Proteins/pharmacokinetics , Administration, Cutaneous , Amino Acid Sequence , Animals , BALB 3T3 Cells , Epidermal Growth Factor/chemistry , Humans , Male , Mice , Peptides/administration & dosage , Peptides/chemistry , Peptides/pharmacokinetics , Rats, Sprague-Dawley , Recombinant Fusion Proteins/chemistry , Skin Absorption
13.
J Pharm Sci ; 102(11): 4109-20, 2013 Nov.
Article in English | MEDLINE | ID: mdl-24018779

ABSTRACT

We evaluated the laser induced burn wound healing efficacy of a recombinant low-molecular-weight protamine conjugated epidermal growth factor (rLMWP-EGF). rLMWP-EGF was prepared by genetically combining LMWP with the N-terminal sequence of EGF; we obtained a homogeneous modified EGF without reduced biological activity. Because of the protein transduction domain of LMWP, rLMWP-EGF showed enhanced drug penetration across artificial skin constructs and excised mouse skin layers versus EGF and showed significantly improved burn wound healing efficacy, with accelerated wound closure and minimized eschar and scar formation, compared with EGF or no treatment. Histological examination also revealed that rLMWP-EGF permeated through the intact skin around the wound and facilitated residual epithelial cell proliferation in an integrated manner to reform an intact epidermis. Radiofrequency microwound formation was effective for reducing large hypertrophic scars formed after severe laser burning by collagen remodeling but rLMWP-EGF did not show a meaningful synergistic effect in burn scar reduction. However, rLMWP-EGF was helpful for forming skin with a more normal appearance and texture. Thus, rLMWP-EGF demonstrated therapeutic potential as a novel topical burn wound healing drug with no obvious toxic effect.


Subject(s)
Burns/drug therapy , Epidermal Growth Factor/therapeutic use , Protamines/therapeutic use , Recombinant Fusion Proteins/therapeutic use , Animals , Burns/pathology , Cell Proliferation/drug effects , Epidermal Growth Factor/administration & dosage , Epidermal Growth Factor/pharmacokinetics , Female , Humans , Male , Mice , Mice, Inbred BALB C , Protamines/administration & dosage , Protamines/pharmacokinetics , Rabbits , Recombinant Fusion Proteins/administration & dosage , Recombinant Fusion Proteins/pharmacokinetics , Skin/drug effects , Skin/metabolism , Skin/pathology , Skin Absorption , Wound Healing/drug effects
14.
Mol Oncol ; 7(3): 475-83, 2013 Jun.
Article in English | MEDLINE | ID: mdl-23298730

ABSTRACT

Targeted toxin-based therapeutics are hindered by poor intracellular uptake, limited stability and non-specific immune stimulation. To address these problems, ligand-targeted toxins in combination with low dose saponin mixtures have been adapted and tested in vivo in the past, however, undefined saponin raw mixtures are not suitable for use in clinical development. In the present work we therefore used a targeted toxin (Sap3-EGF, i.e. saporin fused to epidermal growth factor) in combination with a structurally defined isolated saponin m/z 1861 (SO-1861). In vitro evaluation confirmed a 6900-fold enhancement in the cytotoxic efficacy of Sap3-EGF against TSA-EGFR target cells. The required dose of the targeted toxin was appreciably reduced and there was a highly synergistic effect observed. An ex vivo hemolysis assay showed no or very less hemolysis up to 10 µg/mL of SO-1861. In the acute toxicity studies SO-1861 was found to be non-toxic up to a dose of 100 µg/treatment. The enzymes aspartate aminotransferase, alanine aminotransferase, and glutamate dehydrogenase did not show any statistically significant liver damage, which was further confirmed by histological examination. Additionally, creatinine was also similar to the control group thus ruling out damage to kidney. In vivo studies in a syngeneic BALB/c tumor model characterized by EGFR overexpression were done by applying 30 µg SO-1861 and 0.1 µg Sap3-EGF per treatment. A more than 90% reduction (p < 0.05) in the average tumor volume was observed by this combined therapy.


Subject(s)
Adenocarcinoma/drug therapy , Breast Neoplasms/drug therapy , Drug Delivery Systems , Epidermal Growth Factor/therapeutic use , Immunotoxins/therapeutic use , Pharmaceutical Vehicles/metabolism , Saponins/metabolism , Adenocarcinoma/metabolism , Adenocarcinoma/pathology , Animals , Breast/drug effects , Breast/metabolism , Breast/pathology , Breast Neoplasms/metabolism , Breast Neoplasms/pathology , Cell Line, Tumor , Epidermal Growth Factor/metabolism , Epidermal Growth Factor/pharmacokinetics , ErbB Receptors/metabolism , Female , Humans , Immunotoxins/metabolism , Immunotoxins/pharmacokinetics , Mice , Mice, Inbred BALB C , NIH 3T3 Cells , Pharmaceutical Vehicles/chemistry , Pharmaceutical Vehicles/isolation & purification , Saponaria/chemistry , Saponins/chemistry , Saponins/isolation & purification
15.
Int J Nanomedicine ; 7: 4625-35, 2012.
Article in English | MEDLINE | ID: mdl-22942644

ABSTRACT

Epidermal growth factor receptor (EGFR)-targeted gene delivery is a promising approach in gene therapy against EGFR-positive cancer. In addition, macromolecules, such as polyamidoamine (PAMAM) dendrimers, are potential nonviral gene carriers in this therapy because of their biocompatibility and modifiable features. To achieve the goal of selectively enhancing the transfection efficiency in EGFR-positive cancer cells, the researchers developed chemical approaches of EGF-dendrimer conjugate, which were effective but complicated. Studies on liposomes reveal that self-assembly is another effective but simpler approach in EGF modification. Moreover, properly activated PAMAM dendrimers exhibit higher transfection efficiency, but little research has been done on its ligand-modification. In this study, we developed and characterized a novel gene-delivery system based on activated EGF-dendriplexes, which is formed via self-assembly by EGF and complexes prepared by activated PAMAM dendrimer and plasmid DNA. Such complexes exhibit desired features compared to nonmodified or non-activated dendriplexes in vitro, including selective enhancement of transfection efficiency in EGFR-positive cells, decreased cytotoxicity, and low agonist effect. In vivo experimentation shows their EGFR-positive tumor targeted biodistribution and increased transfection efficiency at EGFR-positive tumors. Our results demonstrated that activated EGF-dendriplexes are safe and effective carriers for delivering gene drugs to EGFR-positive cells, which makes these complexes a promising targeted nonviral gene-delivery system for auxiliary cancer therapy.


Subject(s)
DNA/chemistry , Dendrimers/chemistry , Drug Carriers/chemistry , Epidermal Growth Factor/chemistry , ErbB Receptors/metabolism , Transfection/methods , Animals , Cell Proliferation/drug effects , Cell Survival/drug effects , DNA/pharmacokinetics , Dendrimers/pharmacokinetics , Dendrimers/pharmacology , Drug Carriers/pharmacokinetics , Drug Carriers/pharmacology , Drug Stability , Epidermal Growth Factor/pharmacokinetics , Epidermal Growth Factor/pharmacology , Female , Green Fluorescent Proteins/chemistry , Green Fluorescent Proteins/genetics , Green Fluorescent Proteins/metabolism , HEK293 Cells , Humans , MCF-7 Cells , Mice , Mice, Inbred BALB C , Particle Size , Spectroscopy, Near-Infrared
16.
Nucl Med Biol ; 39(8): 1122-7, 2012 Nov.
Article in English | MEDLINE | ID: mdl-22819251

ABSTRACT

INTRODUCTION: As direct radiolabeled peptides suffer limitations for in vivo imaging, we investigated the usefulness of radioloabeled avidin and streptavidin as cores to link peptide ligands for targeted tumor imaging. METHODS: Human epidermal growth factor (EGF) was site specifically conjugated with a single PEG-biotin molecule and linked to (99m)Tc-HYNIC labeled avidin-FITC (Av) or streptavidin-Cy5.5 (Sav). Receptor targeting was verified in vitro, and in vivo pharmacokinetic and biodistribution profiles were studied in normal mice. Scintigraphic imaging was performed in MDA-MB-468 breast tumor xenografted nude mice. RESULTS: Whereas both (99m)Tc-Av-EGF and (99m)Tc-Sav-EGF retained receptor-specific binding in vitro, the two probes substantially diverged in pharmacokinetic and biodistribution behavior in vivo. (99m)Tc-Av-EGF was rapidly eliminated from the circulation with a T1/2 of 4.3 min, and showed intense hepatic accumulation but poor tumor uptake (0.6%ID/gm at 4 h). (99m)Tc-Sav-EGF displayed favorable in vivo profiles of longer circulation (T1/2ß, 51.5 min) and lower nonspecific uptake that resulted in higher tumor uptake (3.8 %ID/gm) and clear tumor visualization at 15 h. CONCLUSION: (99m)Tc-HYNIC labeled streptavidin linked with growth factor peptides may be useful as a protein-ligand complex for targeted imaging of tumor receptors.


Subject(s)
Avidin/metabolism , Biotin/metabolism , Breast Neoplasms/pathology , Diagnostic Imaging/methods , Epidermal Growth Factor , ErbB Receptors/metabolism , Organotechnetium Compounds , Streptavidin/metabolism , Animals , Avidin/chemistry , Breast Neoplasms/diagnostic imaging , Cell Line, Tumor , Cell Transformation, Neoplastic , Epidermal Growth Factor/chemistry , Epidermal Growth Factor/metabolism , Epidermal Growth Factor/pharmacokinetics , Humans , Mice , Models, Molecular , Organotechnetium Compounds/chemistry , Organotechnetium Compounds/metabolism , Organotechnetium Compounds/pharmacokinetics , Polyethylene Glycols/chemistry , Protein Conformation , Radionuclide Imaging , Streptavidin/chemistry
17.
Pharm Res ; 29(7): 2017-29, 2012 Jul.
Article in English | MEDLINE | ID: mdl-22399389

ABSTRACT

PURPOSE: To evaluate the ability of a novel radiofrequency (RF) microporation technology based on ablation of the skin barrier to enhance topical delivery of active ingredients METHODS: The influence of RF fluence and the molecular size of the absorbent on the permeation enhancement was confirmed by in vitro skin permeation study using Franz diffusion cells. The improved skin rejuvenation effects, such as depigmentation and anti-wrinkle effects, by enhanced topical delivery of α-bisabolol and epidermal growth factor (EGF) through the RF microchannels were investigated in photo-damaged skin. RESULTS: The cumulative amounts of active ingredients through the RF microporated skin were significantly increased. Topically applied α-bisabolol after RF microporation induced rapid onset of skin whitening and significantly increased the ΔL-value of UVB-induced hyperpigmented melanin hairless mouse skin. In addition, wrinkle formation after topical application of EGF with RF microporation was significantly reduced and prevented after 12 weeks, and all parameters involving wrinkles in a replica analysis were similar to those in the negative control. CONCLUSIONS: RF microporation enhances the topical delivery of active ingredients with high molecular weight or of small hydrophilic or lipophilic molecules. Thus, this technology can effectively improve photo-induced hyperpigmentation and wrinkle formation by enhancing topical delivery of active agents.


Subject(s)
Drug Delivery Systems/methods , Epidermal Growth Factor/administration & dosage , Sesquiterpenes/administration & dosage , Skin/metabolism , Administration, Cutaneous , Animals , Epidermal Growth Factor/pharmacokinetics , Epidermal Growth Factor/pharmacology , Mice , Mice, Hairless , Monocyclic Sesquiterpenes , Radio Waves , Sesquiterpenes/pharmacokinetics , Sesquiterpenes/pharmacology , Skin/drug effects , Skin/ultrastructure , Skin Absorption , Skin Aging/drug effects , Skin Pigmentation/drug effects
19.
Int J Radiat Oncol Biol Phys ; 83(4): 1298-305, 2012 Jul 15.
Article in English | MEDLINE | ID: mdl-22336201

ABSTRACT

PURPOSE: The open structure of euchromatin renders it susceptible to DNA damage by ionizing radiation (IR) compared with compact heterochromatin. The effect of chromatin configuration on the efficacy of Auger electron radiotherapy was investigated. METHODS AND MATERIALS: Chromatin structure was altered in MDA-MB-468 and 231-H2N human breast cancer cells by suberoylanilide hydroxamic acid (SAHA), 5-aza-2-deoxycytidine, or hypertonic treatment. The extent and duration of chromatin structural changes were evaluated using the micrococcal nuclease assay. DNA damage (γH2AX assay) and clonogenic survival were evaluated after exposure to (111)In-DTPA-hEGF, an Auger electron-emitting radiopharmaceutical, or IR. The intracellular distribution of (111)In-DTPA-hEGF after chromatin modification was investigated in cell fractionation experiments. RESULTS: Chromatin remained condensed for up to 20 minutes after NaCl and in a relaxed state 24 hours after SAHA treatment. The number of γH2AX foci per cell was greater in MDA-MB-468 and 231-H2N cells after IR (0.5 Gy) plus SAHA (1 µM) compared with IR alone (16 ± 0.6 and 14 ± 0.3 vs. 12 ± 0.4 and 11 ± 0.2, respectively). More γH2AX foci were observed in MDA-MB-468 and 231-H2N cells exposed to (111)In-DTPA-hEGF (6 MBq/µg) plus SAHA vs. (111)In-DTPA-hEGF alone (11 ± 0.3 and 12 ± 0.7 vs. 9 ± 0.4 and 7 ± 0.3, respectively). 5-aza-2-deoxycytidine enhanced the DNA damage caused by IR and (111)In-DTPA-hEGF. Clonogenic survival was reduced in MDA-MB-468 and 231-H2N cells after IR (6 Gy) plus SAHA (1 µM) vs. IR alone (0.6% ± 0.01 and 0.3% ± 0.2 vs. 5.8% ± 0.2 and 2% ± 0.1, respectively) and after (111)In-DTPA-hEGF plus SAHA compared to (111)In-DTPA-hEGF alone (21% ± 0.4% and 19% ± 4.6 vs. 33% ± 2.3 and 32% ± 3.7). SAHA did not affect (111)In-DTPA-hEGF nuclear localization. Hypertonic treatment resulted in fewer γH2AX foci per cell after IR and (111)In-DTPA-hEGF compared to controls but did not significantly alter clonogenic survival. CONCLUSIONS: Chromatin structure affects DNA damage and cell survival after exposure to Auger electron radiation.


Subject(s)
Chromatin/radiation effects , DNA Damage , Epidermal Growth Factor/pharmacology , Pentetic Acid/analogs & derivatives , Radiation Tolerance , Radiopharmaceuticals/pharmacology , Analysis of Variance , Azacitidine/analogs & derivatives , Azacitidine/pharmacology , Cell Line, Tumor , Chromatin/chemistry , Chromatin/drug effects , Chromatin/metabolism , DNA Damage/genetics , Decitabine , Electrons , Epidermal Growth Factor/pharmacokinetics , ErbB Receptors/metabolism , Histone Deacetylase Inhibitors/pharmacology , Histones/analysis , Humans , Hydroxamic Acids/pharmacology , Pentetic Acid/pharmacokinetics , Pentetic Acid/pharmacology , Radiopharmaceuticals/pharmacokinetics , Radiotherapy , Sodium Chloride/pharmacology , Vorinostat
20.
J Control Release ; 157(3): 366-74, 2012 Feb 10.
Article in English | MEDLINE | ID: mdl-21982897

ABSTRACT

The physico-chemical characteristics and composition of block copolymer micelles (BCMs) may influence the pharmacokinetics and consequently, the desired delivery characteristics. In this study the influence of formulation variables such as size, density of targeting ligand [i.e. epidermal growth factor (hEGF)] and the bifunctional chelator (BFC) used for labelling the BCMs with (111)In, on the pharmacokinetics and biodistribution in mice were evaluated. BCMs were prepared from Me-PEG(x)-b-PCL(y) (x=2.5 k, y=1.2 k for 15 nm BCMs and x=5 k, y=5 k for 60 nm BCMs) with (targeted, 1 or 5 mol% hEGF) or without (non-targeted) hEGF-PEG(x)-b-PCL(y). To investigate the effect of the BFC on the pharmacokinetics, the BCMs were labelled with (111)In using p-SCN-Bn-DOTA (Bn-DOTA-PEG(x)-b-PCL(y)), H(2)N-DOTA (DOTA-PEG(x)-b-PCL(y)), DTPA anhydride (DTPA-PEG(x)-b-PCL(y)) or p-SCN-Bn-DTPA (Bn-DTPA-PEG(x)-b-PCL(y)). The resulting 15 nm or 60 nm non-targeted or targeted (1 or 5 mol% hEGF) were injected via a tail vein to mice bearing MDA-MB-468 human breast cancer xenograft that overexpress EGFR, followed by pharmacokinetics and biodistribution studies. Pharmacokinetic parameters were determined by fitting the blood concentration vs time data using a two compartment model with i.v. bolus input. Pharmacokinetic parameters were found to depend on BCM size, the BFC used as well as the density of hEGF on the surface of the BCMs. BCMs labelled with p-SCN-Bn-DTPA ((111)In-Bn-BCMs) showed improved pharmacokinetics (i.e. extended circulation lifetime) and tumor uptake compared to those labelled with DOTA-PEG(x)-b-PCL(y), p-SCN-Bn-DOTA or DTPA dianhydride. Formulations with a high density of hEGF (5 mol% hEGF) had short circulation half-lives. BCMs labelled with (111)In via p-SCN-Bn-DTPA showed highest accumulation in the liver and spleen and slower whole body elimination. Smaller sized BCMs were rapidly cleared from the circulation. Increasing the density of hEGF on the surface did not improve tumor uptake due to faster clearance from the circulation. To achieve improved pharmacokinetics and in turn effective exploitation of the EPR effect, p-SCN-Bn-DTPA emerged as the optimal BFC for radiolabelling BCMs while a lower density of hEGF gave more favourable organ distribution.


Subject(s)
Breast Neoplasms/metabolism , Chelating Agents/pharmacokinetics , Epidermal Growth Factor/pharmacokinetics , Micelles , Polymers/pharmacokinetics , Animals , Cell Line, Tumor , Chelating Agents/chemistry , Chemistry, Pharmaceutical , Epidermal Growth Factor/chemistry , Heterocyclic Compounds, 1-Ring/chemistry , Heterocyclic Compounds, 1-Ring/pharmacokinetics , Humans , Mice , Mice, Nude , Neoplasm Transplantation , Pentetic Acid/chemistry , Pentetic Acid/pharmacokinetics , Tissue Distribution
SELECTION OF CITATIONS
SEARCH DETAIL
...