Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 92
Filter
Add more filters










Publication year range
1.
Pharmaceutics ; 13(8)2021 Jul 27.
Article in English | MEDLINE | ID: mdl-34452106

ABSTRACT

Fluorescently labeled nanoparticles are widely used for evaluating their distribution in the biological environment. However, dye leakage can lead to misinterpretations of the nanoparticles' biodistribution. To better understand the interactions of dyes and nanoparticles and their biological environment, we explored PLGA nanoparticles labeled with four widely used dyes encapsulated (coumarin 6, rhodamine 123, DiI) or bound covalently to the polymer (Cy5.5.). The DiI label was stable in both aqueous and lipophilic environments, whereas the quick release of coumarin 6 was observed in model media containing albumin (42%) or liposomes (62%), which could be explained by the different affinity of these dyes to the polymer and lipophilic structures and which we also confirmed by computational modeling (log PDPPC/PLGA: DiI-2.3, Cou6-0.7). The importance of these factors was demonstrated by in vivo neuroimaging (ICON) of the rat retina using double-labeled Cy5.5/Cou6-nanoparticles: encapsulated Cou6 quickly leaked into the tissue, whereas the stably bound Cy.5.5 label remained associated with the vessels. This observation is a good example of the possible misinterpretation of imaging results because the coumarin 6 distribution creates the impression that nanoparticles effectively crossed the blood-retina barrier, whereas in fact no signal from the core material was found beyond the blood vessels.

2.
Expert Opin Drug Deliv ; 17(1): 23-32, 2020 01.
Article in English | MEDLINE | ID: mdl-31774000

ABSTRACT

Introduction: Current therapies of neurodegenerative or neurometabolic diseases are, to a large extent, hampered by the inability of drugs to cross the blood-brain barrier (BBB). This very tight barrier severely restricts the entrance of molecules from the blood into the brain, especially macromolecular substances (i.e. neurotrophic factors, enzymes, proteins, as well as genetic materials). Due to their size, physicochemical properties, and instability, the delivery of these materials is particularly difficult.Areas covered: Recent research showed that biocompatible and biodegradable nanoparticles possessing tailored surface properties can enable a delivery of drugs and specifically of macromolecules across the blood-brain barrier by using carrier systems of the brain capillary endothelium (Trojan Horse strategy). In the present review, the state-of-art of nanoparticle-mediated drug delivery of different macromolecular substances into the brain following intravenous injection is summarized, and different nanomedicines that are used to enable the transport of neurotrophic factors and enzymes across the blood-brain barrier into the CNS are critically analyzed.Expert opinion: Brain delivery of macromolecules by an intravenous application using nanomedicines is now a growing area of interest which could be really translated into clinical application if dedicated effort will be given to industrial scale-up production.


Subject(s)
Blood-Brain Barrier/metabolism , Brain/metabolism , Drug Carriers/chemistry , Macromolecular Substances/metabolism , Nanoparticles/chemistry , Absorbable Implants , Animals , Biocompatible Materials , Biological Transport , Brain-Derived Neurotrophic Factor/metabolism , Drug Delivery Systems , Enzyme Replacement Therapy , Glial Cell Line-Derived Neurotrophic Factor/metabolism , Humans , Injections, Intravenous , Nanomedicine , Nerve Growth Factor/metabolism
3.
Int J Pharm ; 572: 118733, 2019 Dec 15.
Article in English | MEDLINE | ID: mdl-31689481

ABSTRACT

Brain delivery of drugs by nanoparticles is a promising strategy that could open up new possibilities for the chemotherapy of brain tumors. As demonstrated in previous studies, the loading of doxorubicin in poly(lactide-co-glycolide) nanoparticles coated with poloxamer 188 (Dox-PLGA) enabled the brain delivery of this cytostatic that normally cannot penetrate across the blood-brain barrier in free form. The Dox-PLGA nanoparticles produced a very considerable anti-tumor effect against the intracranial 101.8 glioblastoma in rats, thus representing a promising candidate for the chemotherapy of brain tumors that warrants clinical evaluation. The objective of the present study, therefore, was the optimization of the Dox-PLGA formulation and the development of a pilot scale manufacturing process. Optimization of the preparation procedure involved the alteration of the technological parameters such as replacement of the particle stabilizer PVA 30-70 kDa with a presumably safer low molecular mass PVA 9-10 kDa as well as the modification of the external emulsion medium and the homogenization conditions. The optimized procedure enabled an increase of the encapsulation efficiency from 66% to >90% and reduction of the nanoparticle size from 250 nm to 110 nm thus enabling the sterilization by membrane filtration. The pilot scale process was characterized by an excellent reproducibility with very low inter-batch variations. The in vitro hematotoxicity of the nanoparticles was negligible at therapeutically relevant concentrations. The anti-tumor efficacy of the optimized formulation and the ability of the nanoparticles to penetrate into the intracranial tumor and normal brain tissue were confirmed by in vivo experiments.


Subject(s)
Antibiotics, Antineoplastic/administration & dosage , Brain Neoplasms/drug therapy , Doxorubicin/administration & dosage , Glioblastoma/drug therapy , Nanoparticles/administration & dosage , Polylactic Acid-Polyglycolic Acid Copolymer/administration & dosage , Animals , Antibiotics, Antineoplastic/chemistry , Antibiotics, Antineoplastic/radiation effects , Doxorubicin/chemistry , Doxorubicin/radiation effects , Drug Development , Drug Stability , Male , Nanoparticles/chemistry , Nanoparticles/radiation effects , Polylactic Acid-Polyglycolic Acid Copolymer/chemistry , Polylactic Acid-Polyglycolic Acid Copolymer/radiation effects , Rats, Wistar , Sterilization
4.
Pharmaceutics ; 11(8)2019 Aug 12.
Article in English | MEDLINE | ID: mdl-31409024

ABSTRACT

Human serum albumin nanoparticles (HSA-NPs) have been widely used as drug delivery systems. In most cases, HSA-NPs are formed by the method of desolvation in the presence of glutaraldehyde as a crosslinking agent. In the present study, we showed the possibility of crosslinking human serum albumin (HSA) molecules with natural agents, urea, and cysteine at the nanoparticle level under mild conditions (at room temperature of 20-25 °C). Optimal concentrations of the interacting components (HSA, urea, and cysteine) were found to produce nanoparticles with optimal physico-chemical parameters (particle size, polydispersity, zeta potential, yield, etc.) for application as drug carriers. We used hydroxyurea (HU), a simple organic compound currently used as a cancer chemotherapeutic agent. The results indicated sizes of 196 ± 5 nm and 288 ± 10 nm with a surface charge of -22 ± 3.4 mV and -17.4 ± 0.5 mV for HSA-NPs (20 mg/mL of HSA, 0.01 mg/mL of cysteine, and 10 mg/mL of urea) and HSA-HU-NPs (2 mg/mL of HU), respectively. The yield of the HSA-HU-NPs was ~93% with an encapsulation efficiency of ~77%. Thus, the particles created (immobilized with HU) were stable over time and able to prolong the effect of the drug.

5.
Int J Pharm ; 554: 161-178, 2019 Jan 10.
Article in English | MEDLINE | ID: mdl-30414476

ABSTRACT

Doxorubicin loaded in poloxamer 188-coated PLGA nanoparticles (Dox-NP + P188) was shown to produce a high antitumor effect against the experimental orthotopic 101.8 glioblastoma in rats upon intravenous administration. The objective of the present study was to evaluate the acute and chronic toxicity of this nanoformulation. The parent drug was used as a reference formulation. Acute toxicity of doxorubicin-loaded nanoparticles in mice and rats was similar to that of free doxorubicin. The chronic toxicity study was conducted in Chinchilla rabbits; the treatment regimen consisted of 30 daily intravenous injections using two dosage levels: 0.22 mg/kg/day and 0.15 mg/kg/day. The study included assessment of the body weight, hematological parameters, blood biochemical parameters, urinalysis, and pathomorphological evaluation of the internal organs. The results of the study demonstrated that the hematological, cardiac, and testicular toxicity of doxorubicin could be reduced by binding the drug to PLGA nanoparticles. Coating of PLGA nanoparticles with poloxamer 188 contributed to the reduction of cardiotoxicity. Functional and morphological abnormalities caused by the nanoparticulate doxorubicin were dose-dependent and reversible. Altogether these results provide evidence that the PLGA-based nanoformulation not only might enable the broadening of the spectrum of doxorubicin activity but also an improvement of its safety profile.


Subject(s)
Antibiotics, Antineoplastic/administration & dosage , Doxorubicin/administration & dosage , Glioblastoma/drug therapy , Nanoparticles , Animals , Antibiotics, Antineoplastic/pharmacology , Antibiotics, Antineoplastic/toxicity , Cardiotoxicity/etiology , Dose-Response Relationship, Drug , Doxorubicin/pharmacology , Doxorubicin/toxicity , Drug Carriers/chemistry , Female , Injections, Intravenous , Male , Mice , Poloxamer/chemistry , Polylactic Acid-Polyglycolic Acid Copolymer/chemistry , Rabbits , Rats , Toxicity Tests, Acute , Toxicity Tests, Chronic
6.
Int J Pharm ; 547(1-2): 10-23, 2018 Aug 25.
Article in English | MEDLINE | ID: mdl-29751140

ABSTRACT

Resistance to antiepileptic drugs (AEDs) is a major clinical problem. The overexpression of P-glycoprotein (Pgp), one of the main transporters limiting the entry of xenobiotics into the brain, is among the factors contributing to the AED resistance. Presently, there is no consensus on the interaction of carbamazepine (CBZ) with the Pgp. This study investigates the effect of the Pgp inhibitor verapamil on the anticonvulsant effect of CBZ and its nanoparticulate formulation in the rat model of isoniazid-induced epilepsy. Verapamil significantly increased the anticonvulsant effect of CBZ and reduced its effective dose by at least 30% (from 30 mg/kg to 20 mg/kg). Binding of carbamazepine to the poloxamer 188-coated PLGA nanoparticles enabled a 30-fold increase of its anticonvulsive effect, as compared to the free drug. The inhibition of Pgp did not influence the effectivity of carbamazepine encapsulated in nanoparticles.


Subject(s)
Anticonvulsants/administration & dosage , Carbamazepine/administration & dosage , Drug Resistant Epilepsy/drug therapy , Nanoparticles/administration & dosage , ATP Binding Cassette Transporter, Subfamily B, Member 1/antagonists & inhibitors , Animals , Anticonvulsants/chemistry , Anticonvulsants/pharmacokinetics , Anticonvulsants/therapeutic use , Brain/physiopathology , Carbamazepine/chemistry , Carbamazepine/pharmacokinetics , Carbamazepine/therapeutic use , Dose-Response Relationship, Drug , Drug Delivery Systems , Drug Resistant Epilepsy/chemically induced , Drug Resistant Epilepsy/physiopathology , Electrocorticography , Isoniazid , Lactic Acid/administration & dosage , Lactic Acid/chemistry , Lactic Acid/pharmacokinetics , Lactic Acid/therapeutic use , Male , Nanoparticles/chemistry , Nanoparticles/therapeutic use , Poloxamer/administration & dosage , Poloxamer/chemistry , Poloxamer/pharmacokinetics , Poloxamer/therapeutic use , Polyglycolic Acid/administration & dosage , Polyglycolic Acid/chemistry , Polyglycolic Acid/pharmacokinetics , Polyglycolic Acid/therapeutic use , Polylactic Acid-Polyglycolic Acid Copolymer , Rats, Wistar , Verapamil/pharmacology
7.
Int J Pharm ; 524(1-2): 77-90, 2017 May 30.
Article in English | MEDLINE | ID: mdl-28359811

ABSTRACT

The paramount problem in the therapy of brain tumors is the inability of most drugs to cross the blood-brain barrier. PLGA nanoparticles overcoated with poloxamer 188 could overcome this problem and enabled a high anti-tumoral effect against the very aggressive intracranial 101.8 glioblastoma in rats that closely resembles human grade IV glioblastomas. The basis for the transport of these particles across the blood-brain barrier appears to be adsorption of blood apolipoproteins (ApoE or ApoA-I) on the nanoparticle surface caused by the poloxamer 188-coating, followed by receptor-mediated transcytosis of the nanoparticles. The objective of the present study is the elucidation of the mechanism by which the poloxamer 188-coated nanoparticles then enter the brain tumor cells. Their intracellular fate, therefore, was investigated using the U87 human glioma cell line. The main mechanism of the PLGA nanoparticle internalization by U87 cells was clathrin-mediated endocytosis. Within 1h free doxorubicin was released from late endosomes and could reach its target site, i.e. the DNA in the nuclei without degradation, whereas the PLGA nanoparticles, which were labeled with Cy5.5, still were observed in the endo-lysosomal compartment. These results demonstrate that the underlying mechanism of action in the brain cells is by diffusive doxorubicin release from the nanoparticles rather than by their intracellular degradation.


Subject(s)
Doxorubicin/administration & dosage , Glioblastoma/drug therapy , Lactic Acid/chemistry , Nanoparticles/chemistry , Polyglycolic Acid/chemistry , Blood-Brain Barrier , Cell Line, Tumor , Drug Liberation , Humans , Polylactic Acid-Polyglycolic Acid Copolymer
8.
J Control Release ; 253: 1-10, 2017 05 10.
Article in English | MEDLINE | ID: mdl-28215668

ABSTRACT

The lysosomal storage disorder (LSD) metachromatic leukodystrophy (MLD) is caused by a deficiency of the soluble, lysosomal hydrolase arylsulfatase A (ASA). The disease is characterized by accumulation of 3-O-sulfogalactosylceramide (sulfatide), progressive demyelination of the nervous system and premature death. Enzyme replacement therapy (ERT), based on regular intravenous injections of recombinant functional enzyme, is in clinical use for several LSDs. For MLD and other LSDs with central nervous system (CNS) involvement, however, ERT is limited by the blood-brain barrier (BBB) restricting transport of therapeutic enzymes from the blood to the brain. In the present study, the potential of different types of surfactant-coated biodegradable nanoparticles to increase brain delivery of ASA was evaluated. Three different strategies to bind ASA to nanoparticle surfaces were compared: (1) adsorption, (2) high-affinity binding via the streptavidin-biotin system, and (3) covalent binding. Adsorption allowed binding of high amounts of active ASA. However, in presence of phosphate-buffered saline or serum rapid and complete desorption occurred, rendering this strategy ineffective for in vivo applications. In contrast, stable immobilization with negligible dissociation was achieved by high-affinity and covalent binding. Consequently, we analyzed the brain targeting of two stably nanoparticle-bound ASA formulations in ASA-/- mice, an animal model of MLD. Compared to free ASA, injected as a control, the biodistribution of nanoparticle-bound ASA was altered in peripheral organs, but no increase of brain levels was detectable. The failure to improve brain delivery suggests that the ASA glycoprotein interferes with processes required to target surfactant-coated nanoparticles to brain capillary endothelial cells.


Subject(s)
Brain/metabolism , Cerebroside-Sulfatase/administration & dosage , Nanoparticles/administration & dosage , Surface-Active Agents/administration & dosage , Animals , Avidin/chemistry , Biotinylation , Cerebroside-Sulfatase/chemistry , Cerebroside-Sulfatase/genetics , Cerebroside-Sulfatase/pharmacokinetics , Female , Lactic Acid/chemistry , Leukodystrophy, Metachromatic/drug therapy , Leukodystrophy, Metachromatic/metabolism , Mice, Knockout , Nanoparticles/chemistry , Poloxamer/administration & dosage , Poloxamer/chemistry , Poloxamer/pharmacokinetics , Polyesters/chemistry , Polyglycolic Acid/chemistry , Polylactic Acid-Polyglycolic Acid Copolymer , Polysorbates/administration & dosage , Polysorbates/chemistry , Polysorbates/pharmacokinetics , Serum Albumin, Human/chemistry , Surface-Active Agents/chemistry , Surface-Active Agents/pharmacokinetics
9.
Drug Deliv ; 23(9): 3520-3528, 2016 Nov.
Article in English | MEDLINE | ID: mdl-27278330

ABSTRACT

Currently, traumatic brain injury (TBI) is the leading cause of death or disabilities in young individuals worldwide. The multi-complexity of its pathogenesis as well as impermeability of the blood-brain barrier (BBB) makes the drug choice and delivery very challenging. The brain-derived neurotrophic factor (BDNF) regulates neuronal plasticity, neuronal cell growth, proliferation, cell survival and long-term memory. However, its short half-life and low BBB permeability are the main hurdles to be an effective therapeutic for TBI. Poly (lactic-co-glycolic acid) (PLGA) nanoparticles coated by surfactant can enable the delivery of a variety of molecules across the BBB by receptor-mediated transcytosis. This study examines the ability of PLGA nanoparticles coated with poloxamer 188 (PX) to deliver BDNF into the brain and neuroprotective effects of BNDF in mice with TBI. C57bl/6 mice were subjected to weight-drop closed head injuries under anesthesia. Using enzyme-linked immunosorbent assay, we demonstrated that the intravenous (IV) injection of nanoparticle-bound BDNF coated by PX (NP-BDNF-PX) significantly increased BDNF levels in the brain of sham-operated mice (p < 0.001) and in both ipsi- (p < 0.001) and contralateral (p < 0.001) parts of brain in TBI mice compared to controls. This study also showed using the passive avoidance (PA) test, that IV injection of NP-BDNF-PX 3 h post-injury prolonged the latent time in mice with TBI thereby reversing cognitive deficits caused by brain trauma. Finally, neurological severity score test demonstrated that our compound efficiently reduced the scores at day 7 after the injury indicating the improvement of neurological deficit in animals with TBI. This study shows that PLGA nanoparticles coated with PX effectively delivered BDNF into the brain, and improved neurological and cognitive deficits in TBI mice, thereby providing a neuroprotective effect.


Subject(s)
Brain Injuries, Traumatic/drug therapy , Brain-Derived Neurotrophic Factor/administration & dosage , Brain/drug effects , Cognition/drug effects , Lactic Acid/chemistry , Nanoparticles/chemistry , Polyglycolic Acid/chemistry , Animals , Blood-Brain Barrier/metabolism , Brain-Derived Neurotrophic Factor/chemistry , Half-Life , Male , Mice , Mice, Inbred C57BL , Nanoparticles/administration & dosage , Neuroprotective Agents/administration & dosage , Neuroprotective Agents/chemistry , Poloxamer/chemistry , Polylactic Acid-Polyglycolic Acid Copolymer , Surface-Active Agents/chemistry
10.
Pharmaceutics ; 7(1): 3-9, 2015 Feb 03.
Article in English | MEDLINE | ID: mdl-25654637

ABSTRACT

Little attention so-far has been paid to the influence of chronobiology on the processes of nanoparticle uptake and transport into the brain, even though this transport appears to be chronobiologically controlled to a significant degree. Nanoparticles with specific surface properties enable the transport across the blood-brain barrier of many drugs that normally cannot cross this barrier. A clear dependence of the central antinociceptive (analgesic) effects of a nanoparticle-bound model drug, i.e., the hexapeptide dalargin, on the time of day was observable after intravenous injection in mice. In addition to the strongly enhanced antinociceptive effect due to the binding to the nanoparticles, the minima and maxima of the pain reaction with the nanoparticle-bound drug were shifted by almost half a day compared to the normal circadian nociception: The maximum in the pain reaction after i.v. injection of the nanoparticle-bound dalargin occurred during the later rest phase of the animals whereas the normal pain reaction and that of a dalargin solution was highest during the active phase of the mice in the night. This important shift could be caused by an enhanced endo- and exocytotic particulates transport activity of the brain capillary endothelial cells or within the brain during the rest phase.

11.
Nanotechnology ; 26(4): 045601, 2015 Jan 30.
Article in English | MEDLINE | ID: mdl-25556693

ABSTRACT

Human serum albumin (HSA) is a protein found in human blood. Over the last decade, HSA has been evaluated as a promising drug carrier. However, not being magnetic, HSA cannot be used for biomedical applications such as magnetic resonance imaging (MRI) and magnetic drug targeting. Therefore, subsequent composites building on iron oxide nanoparticles that are already used clinically as MRI contrast agents are extensively studied. Recently and in this context, innovative fully hydrophilic ultra-small CAN-stabilized maghemite ((CeLn)(3/4+)-γ-Fe2O3) nanoparticles have been readily fabricated. The present study discusses the design, fabrication, and characterization of a dual phase hybrid core (rHSA)-shell ((CeLn)(3/4+)-γ-Fe2O3 NPs) nanosystem. Quite importantly and in contrast to widely used encapsulation strategies, rHSA NP surface-attached (CeLn)(3/4+)-γ-Fe2O3 NPs enabled to exploit both rHSA (protein functionalities) and (CeLn)(3/4+)-γ-Fe2O3 NP surface functionalities (COOH and ligand L coordinative exchange) in addition to very effective MRI contrast capability due to optimal accessibility of H2O molecules with the outer magnetic phase. Resulting hybrid nanoparticles might be used as a platform modular system for therapeutic (drug delivery system) and MR diagnostic purposes.


Subject(s)
Biocompatible Materials/chemical synthesis , Magnetite Nanoparticles/chemistry , Serum Albumin/chemistry , Cations , Contrast Media/chemical synthesis , Humans , Magnetite Nanoparticles/toxicity , Materials Testing
12.
J Control Release ; 199: 63-71, 2015 Feb 10.
Article in English | MEDLINE | ID: mdl-25499552

ABSTRACT

Nanoparticle (NP)-based contrast agents that enable high resolution anatomic T1-weighted magnetic resonance imaging (MRI) offer the prospect of improving differential diagnosis of liver tumors such as hepatocellular carcinoma (HCC). In the present study, we investigated the possibility of employing novel non-toxic human serum albumin nanoparticles conjugated with Gd-DTPA and rhodamine 123 (Gd-Rho-HSA-NPs) for the detection of HCC by T1-weighted MRI. In addition, the influence of surface coating of the NPs with poloxamine 908, which alters the absorptive behavior of NPs and changes their distribution between the liver and tumor was examined. MRI of transgenic mice with endogenously formed HCCs following intravenous injection of Gd-Rho-HSA-NPs revealed a strong negative contrast of the tumors. Contrasting of the HCCs by NP-enhanced MRI required less Gd as compared to gadolinium-ethoxybenzyl-diethylenetriaminepentaacetic acid-enhanced MRI, which currently provides the most sensitive detection of HCC in patients. Immunohistochemical analyses revealed that the Gd-Rho-HSA-NPs were localized to macrophages, which were - similar to HCC in patients - fewer in number in HCC as compared to the liver tissue, which is in agreement with the negative contrasting of HCC in Gd-Rho-HSA-NP-enhanced MRI. Poloxamine-coated NPs showed lower accumulation in the tumor macrophages and caused a longer lasting enhancement of the MRI signal. These data indicate that Gd-Rho-HSA-NPs enable sensitive detection of HCC by T1-weighted MRI in mice with endogenous HCC through their uptake by macrophages. Poloxamine coating of the NPs delayed the tumor localization of the NPs.


Subject(s)
Carcinoma, Hepatocellular/diagnosis , Contrast Media , Gadolinium DTPA , Liver Neoplasms/diagnosis , Magnetic Resonance Imaging/methods , Rhodamine 123 , Serum Albumin , Animals , Carcinoma, Hepatocellular/pathology , Cell Survival/drug effects , Ethylenediamines , Excipients , Genes, myc/genetics , Humans , Liver/pathology , Liver Neoplasms/pathology , Macrophages, Peritoneal/drug effects , Mice , Mice, Transgenic , Nanoparticles , Particle Size , Polyethylene Glycols , Tissue Distribution , Transforming Growth Factor alpha/genetics
13.
PLoS One ; 9(9): e107603, 2014.
Article in English | MEDLINE | ID: mdl-25229502

ABSTRACT

The genetic treatment of neurodegenerative diseases still remains a challenging task since many approaches fail to deliver the therapeutic material in relevant concentrations into the brain. As viral vectors comprise the risk of immune and inflammatory responses, human serum albumin (HSA) nanoparticles were found to represent a safer and more convenient alternative. Their ability to cross the blood-brain barrier (BBB) and deliver drugs into the brain in order to enhance gene-based therapy has been previously demonstrated. The present study deals with the development of pGL3-PEI-coated HSA nanoparticles and subsequent in vitro testing in cerebellar granular and HeLa cells. The luciferase control vector pGL3 was chosen as reporter plasmid encoding for the firefly luciferase protein, linear polyethylenimine (22 kDa) as endosomolytic agent for enhancing the cells' transfection. Studies on particle characteristics, their cellular uptake into aforementioned cell lines and on subcellular localisation, and transfection efficiency in the cerebellar cells proved the feasibility of nanoparticle-based gene delivery.


Subject(s)
Drug Carriers/chemistry , Nanoparticles/chemistry , Polyethyleneimine/chemistry , Serum Albumin/chemistry , Transfection , Animals , Cerebellum/cytology , Cerebellum/metabolism , DNA/chemistry , DNA/genetics , Drug Carriers/metabolism , Endocytosis , HeLa Cells , Humans , Mice , Neurons/metabolism , Particle Size , Plasmids/genetics
14.
J Control Release ; 194: 130-7, 2014 Nov 28.
Article in English | MEDLINE | ID: mdl-25173842

ABSTRACT

Iron oxide-containing magnetic nanoparticles (MNPs) have certain advantages over currently used contrast agents for tumor imaging by magnetic resonance imaging (MRI) as they offer the possibility of functionalization with ligands and tracers. Functionalized MNPs also may be used for targeted tumor therapy. In the current study nanoparticles (NPs) consisting of recombinant human serum albumin (rHSA) with incorporated hydrophilic (NH4)2Ce(IV)(NO3)6-γ-Fe2O3 particles (CAN maghemite particles) for medical imaging were produced and characterized. For this purpose CAN maghemite particles were incorporated into an rHSA matrix to yield rHSA-NPs. The resulting NPs were analyzed by transmission electron microscopy, photon correlation spectroscopy, and atomic absorption. The sizes of the manufactured NP were 170 ± 10 nm, and the zeta-potential was -50 ± 3 mV. The NPs remained stable when stored after lyophilization with sucrose 3% [w/v] as a cryoprotector. They showed pro-inflammatory properties without cell and animal toxicity in vivo and were highly contrasting in MRI. In conclusion, this report introduces novel rHSA NP with favorable properties containing iron oxide for detection by MRI.


Subject(s)
Contrast Media , Diagnostic Imaging/methods , Ferric Compounds , Magnetite Nanoparticles , Serum Albumin , Animals , Cell Line, Tumor , Cell Survival/drug effects , Contrast Media/administration & dosage , Contrast Media/toxicity , Drug Stability , Electrochemistry , Ferric Compounds/administration & dosage , Ferric Compounds/chemistry , Ferric Compounds/toxicity , Humans , Magnetic Resonance Imaging , Magnetics , Magnetite Nanoparticles/administration & dosage , Magnetite Nanoparticles/toxicity , Male , Mice , Mice, Inbred C57BL , Mice, Inbred ICR , Neoplasms/diagnosis , Particle Size , Recombinant Proteins/administration & dosage , Recombinant Proteins/toxicity , Serum Albumin/administration & dosage , Serum Albumin/toxicity
15.
Eur J Pharm Biopharm ; 88(2): 510-7, 2014 Oct.
Article in English | MEDLINE | ID: mdl-24995841

ABSTRACT

Severe intoxications with organophosphates require the immediate administration of atropine in combination with acetyl cholinesterase (AChE) reactivators such as HI-6. Although this therapy regimen enables the treatment of peripheral symptoms, the blood-brain barrier (BBB) restricts the access of the hydrophilic antidotes to the central nervous system which could lead to a fatal respiratory arrest. Therefore, HI-6-loaded albumin nanoparticles were previously developed to enhance the transport across this barrier and were able to reactivate organophosphate-(OP)-inhibited AChE in an in vitro BBB model. Since HI-6 is known to be moisture-sensitive, the feasibility of freeze-drying of the HI-6-loaded nanoparticles was investigated in the present study using different cryo- and lyoprotectants at different concentrations. Trehalose and sucrose (3%, w/v)-containing formulations were superior to mannitol concerning the physicochemical parameters of the nanoparticles whereas trehalose-containing samples were subject of a prolonged storage stability study at temperatures between -20°C and +40°C for predetermined time intervals. Shelf-life computations of the freeze-dried HI-6 nanoparticle formulations revealed a shelf-life time of 18 months when stored at -20°C. The formulations' efficacy was proven in vitro by reactivation of OP-inhibited AChE after transport over a porcine brain capillary endothelial cell layer model.


Subject(s)
Drug Stability , Freeze Drying , Nanoparticles , Serum Albumin/chemistry , Blood-Brain Barrier , Feasibility Studies , Humans , In Vitro Techniques , Microscopy, Electron, Scanning , Recombinant Proteins/chemistry
16.
Adv Drug Deliv Rev ; 71: 2-14, 2014 May.
Article in English | MEDLINE | ID: mdl-23981489

ABSTRACT

Nanoparticles enable the delivery of a great variety of drugs including anticancer drugs, analgesics, anti-Alzheimer's drugs, cardiovascular drugs, protease inhibitors, and several macromolecules into the brain after intravenous injection of animals. The mechanism of the nanoparticle-mediated drug transport across the BBB appears to be receptor-mediated endocytosis followed by transcytosis into the brain or by drug release within the endothelial cells. Modification of the nanoparticle surface with covalently attached targeting ligands or by coating with certain surfactants that lead to the adsorption of specific plasma proteins after injection is necessary for this receptor-mediated uptake. A very critical and important requirement for nanoparticulate brain delivery is that the employed nanoparticles are biocompatible and, moreover, rapidly biodegradable, i.e. over a time frame of a few days. In addition to enabling drug delivery to the brain, nanoparticles, as with doxorubicin, may importantly reduce the drug's toxicity and adverse effects due to an alteration of the body distribution. Because of the possibility to treat severe CNS diseases such as brain tumours and to even transport proteins and other macromolecules across the blood-brain barrier, this technology holds great promise for a non-invasive therapy of these diseases.


Subject(s)
Drug Delivery Systems , Nanoparticles , Polymers/chemistry , Animals , Biological Transport , Blood-Brain Barrier/metabolism , Brain/metabolism , Drug-Related Side Effects and Adverse Reactions/prevention & control , Endocytosis/physiology , Humans , Time Factors , Transcytosis/physiology
17.
Eur J Pharm Biopharm ; 87(1): 132-41, 2014 May.
Article in English | MEDLINE | ID: mdl-24365328

ABSTRACT

Tumor visualization by magnetic resonance imaging (MRI) and nanoparticle-based contrast agents may improve the imaging of solid tumors such as hepatocellular carcinoma (HCC). In particular, human serum albumin (HSA) nanoparticles appear to be a suitable carrier due to their safety and feasibility of functionalization. In the present study HSA nanoparticles were conjugated with gadolinium diethylenetriaminepentaacetic acid (Gd-DTPA) using carbodiimide chemistry. The nanoparticles had a uniform spherical shape and a diameter of 235±19nm. For better optical visualization in vitro and in vivo, the HSA-Gd nanoparticles were additionally labeled with rhodamine 123. As shown by confocal microscopy and flow cytometry analysis, the fluorescent nanoparticles were readily taken up by Huh-7 hepatocellular carcinoma cells. After 24h incubation in blood serum, less than 5% of the Gd(III) was released from the particles, which suggests that this nanoparticulate system may be stable in vivo and, therefore, may serve as potentially safe T1 MRI contrast agent for MRI of hepatocellular carcinoma.


Subject(s)
Albumins/chemistry , Biocompatible Materials/chemistry , Carcinoma, Hepatocellular/diagnosis , Contrast Media/chemistry , Gadolinium DTPA/chemistry , Liver Neoplasms/diagnosis , Magnetic Resonance Imaging , Nanoparticles/chemistry , Albumins/pharmacokinetics , Animals , Biocompatible Materials/adverse effects , Carcinoma, Hepatocellular/pathology , Cell Line, Tumor , Cell Survival/drug effects , Contrast Media/adverse effects , Gadolinium DTPA/pharmacokinetics , Humans , Liver Neoplasms/pathology , Liver Neoplasms, Experimental/diagnosis , Liver Neoplasms, Experimental/pathology , Mice , Mice, Transgenic , Nanoparticles/adverse effects , Particle Size , Serum Albumin/chemistry , Surface Properties
18.
J Control Release ; 172(1): 201-206, 2013 Nov 28.
Article in English | MEDLINE | ID: mdl-24008150

ABSTRACT

Nanoparticles (NP) as carriers for anti-cancer drugs have shown great promise. Specific targeting of NP to malignant cells, however, remains an unsolved problem. Conjugation of antibodies specific for tumor membrane antigens to NP represents one approach to improve specificity and to increase therapeutic efficacy. In the present study, for the first time a novel membrane heat shock protein (Hsp70)-specific antibody (cmHsp70.1) was coupled to human serum albumin (HSA) NP, loaded with microRNA (miRNA) plasmids to target the inhibitor of apoptosis protein survivin. The physicochemical properties of monodisperse miRNA-loaded NP showed a diameter of 180 nm to 220 nm, a plasmid incorporation of more than 95% and a surface binding capacity of the antibody of 70-80%. Antibody-conjugated NP displayed an increased cellular uptake in U87MG and LN229 glioblastoma cells compared to isotype control antibody, PEG-coupled controls and peripheral blood lymphocytes (PBL). Survivin expression was significantly reduced in cells treated with the Hsp70-miRNA-NP as compared to non-conjugated NP. Hsp70-miRNA-NP enhanced radiation-induced increase in caspase 3/7 activity and decrease in clonogenic cell survival. In summary, cmHsp70.1 miRNA-NP comprise an enhanced tumor cell uptake and increased therapeutic efficacy of radiation therapy in vitro and provide the basis for the development of antibody-based advanced carrier systems for a tumor cell specific targeting.


Subject(s)
Antibodies, Immobilized/chemistry , Glioblastoma/genetics , Glioblastoma/therapy , HSP70 Heat-Shock Proteins/immunology , Inhibitor of Apoptosis Proteins/genetics , MicroRNAs/administration & dosage , Nanoparticles/chemistry , Antibodies, Immobilized/immunology , Cell Line, Tumor , Gene Knockdown Techniques , Genetic Therapy , Glioblastoma/pathology , Glioblastoma/radiotherapy , Humans , MicroRNAs/genetics , MicroRNAs/therapeutic use , Nanoparticles/metabolism , Nanoparticles/ultrastructure , Plasmids/administration & dosage , Plasmids/genetics , Plasmids/therapeutic use , Survivin
19.
Eur J Pharm Biopharm ; 85(1): 53-60, 2013 Sep.
Article in English | MEDLINE | ID: mdl-23395970

ABSTRACT

The biodistribution of nanoparticles is significantly influenced by their interaction with plasma proteins. In order to optimize and possibly monitor the delivery of drugs bound to nanoparticles across the blood-brain barrier (BBB), the protein adsorption pattern of uncoated poly(lactic-co-glycolic acid) (PLGA) nanoparticles after their incubation in human plasma was studied by mass spectrometry. After washing of the particles with water, the proteins were directly digested on the nanoparticle surface using trypsin and then analyzed by nLC MALDI-TOF/TOF. Up to now, the standard method for investigation into the plasma protein adsorption to the particles was 2D gel electrophoresis (2D-PAGE), in certain cases followed by mass spectrometry. The non-gel-based method proposed in the present study provides novel insights into the protein corona surrounding the nanoparticles. The proteins adsorbed on the PLGA nanoparticles after incubation that gave the best signal in terms of quality (high MASCOT score) in human plasma were apolipoprotein E, vitronectin, histidine-rich glycoprotein and kininogen-1. These proteins also are constituents of HDL.


Subject(s)
Blood Proteins/chemistry , Drug Carriers , Lactic Acid/chemistry , Nanoparticles/chemistry , Polyglycolic Acid/chemistry , Adsorption , Apolipoproteins E/analysis , Apolipoproteins E/chemistry , Apolipoproteins E/metabolism , Blood Banks , Blood Proteins/analysis , Blood Proteins/metabolism , Chromatography, High Pressure Liquid , Databases, Protein , Drug Carriers/pharmacokinetics , Feasibility Studies , Humans , Kininogens/analysis , Kininogens/chemistry , Kininogens/metabolism , Lipoproteins, HDL/chemistry , Microchemistry , Peptide Mapping , Polylactic Acid-Polyglycolic Acid Copolymer , Proteins/analysis , Proteins/chemistry , Proteins/metabolism , Spectrometry, Mass, Matrix-Assisted Laser Desorption-Ionization , Surface Properties , Tandem Mass Spectrometry , Vitronectin/analysis , Vitronectin/chemistry , Vitronectin/metabolism
20.
J Microencapsul ; 30(1): 49-54, 2013.
Article in English | MEDLINE | ID: mdl-22676632

ABSTRACT

In 1995 it was reported for the first time that nanoparticles could be used for the delivery of drugs across the blood-brain barrier (BBB) following intravenous injection. In vitro and in vivo experiments show that the underlying mechanism is receptor-mediated endocytosis followed by transcytosis. No opening of the tight junctions was observed. Due to the overcoating of the nanoparticles with polysorbate 80 or poloxamers 188, apolipoproteins A-I and/or E are adsorbed from the blood on to the particle surface after injection. These apolipoproteins mediate the interaction with LDL or scavenger receptors on the BBB followed by the above brain uptake processes. Likewise, covalent attachment of these apolipoproteins or of transferrin, insulin or antibodies against the respective receptors also enables a similar nanoparticle-mediated drug transport across the BBB. From these results it can be concluded that the nanoparticles act as "Trojan Horses" taking advantage of physiological receptor-mediated transport processes across the BBB.


Subject(s)
Blood-Brain Barrier , Nanoparticles , Polymers/pharmacokinetics , Animals , Doxorubicin/pharmacokinetics , Mice , Nerve Growth Factor/pharmacokinetics
SELECTION OF CITATIONS
SEARCH DETAIL
...