Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 4 de 4
Filter
Add more filters










Database
Language
Publication year range
1.
Pharmaceutics ; 15(4)2023 Apr 17.
Article in English | MEDLINE | ID: mdl-37111743

ABSTRACT

Pharmacometric analysis is often used to quantify the differences and similarities between formulation prototypes. In the regulatory framework, it plays a significant role in the evaluation of bioequivalence. While non-compartmental analysis provides an unbiased data evaluation, mechanistic compartmental models such as the physiologically-based nanocarrier biopharmaceutics model promise improved sensitivity and resolution for the underlying causes of inequivalence. In the present investigation, both techniques were applied to two nanomaterial-based formulations for intravenous injection, namely, albumin-stabilized rifabutin nanoparticles and rifabutin-loaded PLGA nanoparticles. The antibiotic rifabutin holds great potential for the treatment of severe and acute infections of patients co-infected with human immunodeficiency virus and tuberculosis. The formulations differ significantly in their formulation and material attributes, resulting in an altered biodistribution pattern as confirmed in a biodistribution study in rats. The albumin-stabilized delivery system further undergoes a dose-dependent change in particle size which leads to a small yet significant change in the in vivo performance. A second analysis was conducted comparing the dose fraction-scaled pharmacokinetic profiles of three dose levels of albumin-stabilized rifabutin nanoparticles. The dose strength affects both the nanomaterial-related absorption and biodistribution of the carrier as well as the drug-related distribution and elimination parameters, increasing the background noise and difficulty of detecting inequivalence. Depending on the pharmacokinetic parameter (e.g., AUC, Cmax, Clobs), the relative (percentage) difference from the average observed using non-compartmental modeling ranged from 85% to 5.2%. A change in the formulation type (PLGA nanoparticles vs. albumin-stabilized rifabutin nanoparticles) resulted in a similar level of inequivalence as compared to a change in the dose strength. A mechanistic compartmental analysis using the physiologically-based nanocarrier biopharmaceutics model led to an average difference of 152.46% between the two formulation prototypes. Albumin-stabilized rifabutin nanoparticles tested at different dose levels led to a 128.30% difference, potentially due to changes in particle size. A comparison of different dose strengths of PLGA nanoparticles, on average, led to a 3.87% difference. This study impressively illustrates the superior sensitivity of mechanistic compartmental analysis when dealing with nanomedicines.

2.
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
3.
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
4.
Eur J Pharm Biopharm ; 74(2): 157-63, 2010 Feb.
Article in English | MEDLINE | ID: mdl-19755158

ABSTRACT

Poly(lactide-co-glycolide) (PLGA) nanoparticles coated with poloxamer 188 (Pluronic((R)) F-68) or polysorbate 80 (Tween((R)) 80) enable an efficient brain delivery of the drugs after intravenous injection. This ability was evidenced by two different pharmacological test systems employing as model drugs the anti-tumour antibiotic doxorubicin and the agonist of opioid receptors loperamide, which being P-gp substrates can cross the blood-brain barrier (BBB) only in pharmacologically insignificant amounts: binding of doxorubicin to the surfactant-coated PLGA nanoparticles, however, enabled a high anti-tumour effect against an intracranial 101/8 glioblastoma in rats, and the penetration of nanoparticle-bound loperamide into the brain was demonstrated by the induction of central analgesic effects in mice. Both pharmacological tests could demonstrate that therapeutic amounts of the drugs were delivered to the sites of action in the brain and showed the high efficiency of the surfactant-coated PLGA nanoparticles for brain delivery. The results of the study also demonstrated that the efficacy of brain delivery by nanoparticles not only is influenced by the coating surfactants but also by other formulation parameters such as core polymer, drug, and stabilizer.


Subject(s)
Brain/drug effects , Chemistry, Pharmaceutical/methods , Doxorubicin/pharmacology , Drug Carriers/chemistry , Loperamide/pharmacology , Pain Threshold/drug effects , Polyglactin 910/pharmacology , Surface-Active Agents , Animals , Cell Line, Tumor , Doxorubicin/administration & dosage , Drug Carriers/administration & dosage , Female , Loperamide/administration & dosage , Male , Mice , Mice, Inbred ICR , Nanoparticles/administration & dosage , Nanoparticles/chemistry , Poloxamer/chemistry , Polyglactin 910/chemistry , Polysorbates/chemistry , Random Allocation , Rats , Rats, Wistar , Xenograft Model Antitumor Assays
SELECTION OF CITATIONS
SEARCH DETAIL
...