Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 7 de 7
Filter
Add more filters










Database
Language
Publication year range
1.
J Pharm Sci ; 106(4): 1149-1161, 2017 04.
Article in English | MEDLINE | ID: mdl-28007561

ABSTRACT

Actively loaded liposomal formulations of anticancer agents have been widely explored due to their high drug encapsulation efficiencies and prolonged drug retention. Mathematical models to predict and optimize drug loading and release kinetics from these nanoparticle formulations would be useful in their development and may allow researchers to tune release profiles. Such models must account for the driving forces as influenced by the physicochemical properties of the drug and the microenvironment, and the liposomal barrier properties. This study employed mechanistic modeling to describe the active liposomal loading and release kinetics of the anticancer agent topotecan (TPT). The model incorporates ammonia transport resulting in generation of a pH gradient, TPT dimerization, TPT lactone ring-opening and -closing interconversion kinetics, chloride transport, and transport of TPT-chloride ion-pairs to describe the active loading and release kinetics of TPT in the presence of varying chloride concentrations. Model-based predictions of the kinetics of active loading at varying loading concentrations of TPT and release under dynamic dialysis conditions were in reasonable agreement with experiments. These findings identify key attributes to consider in optimizing and predicting loading and release of liposomal TPT that may also be applicable to liposomal formulations of other weakly basic pharmaceuticals.


Subject(s)
Models, Chemical , Topotecan/metabolism , Biological Transport/physiology , Liposomes , Topoisomerase I Inhibitors/chemistry , Topoisomerase I Inhibitors/metabolism , Topotecan/chemistry
2.
Sci Rep ; 5: 15236, 2015 Oct 15.
Article in English | MEDLINE | ID: mdl-26468877

ABSTRACT

Low dose metronomic chemotherapy (LDMC) refers to prolonged administration of low dose chemotherapy designed to minimize toxicity and target the tumor endothelium, causing tumor growth inhibition. Topotecan (TPT) when administered at its maximum tolerated dose (MTD) is often associated with systemic hematological toxicities. Liposomal encapsulation of TPT enhances efficacy by shielding it from systemic clearance, allowing greater uptake and extended tissue exposure in tumors. Extended release of TPT from liposomal formulations also has the potential to mimic metronomic therapies with fewer treatments. Here we investigate potential toxicities of equivalent doses of free and actively loaded liposomal TPT (LTPT) and compare them to a fractionated low dose regimen of free TPT in tumor-endothelial spheroids (TES) with/without radiation exposure for a prolonged period of 10 days. Using confocal microscopy, TPT fluorescence was monitored to determine the accumulation of drug within TES. These studies showed TES, being more reflective of the in vivo tumor microenvironment, were more sensitive to LTPT in comparison to free TPT with radiation. More importantly, the response of TES to low-dose metronomic TPT with radiation was comparable to similar treatment with LTPT. This TES study suggests nanoparticle formulations designed for extended release of drug can simulate LDMC in vivo.


Subject(s)
Liposomes/chemistry , Topotecan/chemistry , Animals , Apoptosis/drug effects , Apoptosis/radiation effects , Cell Line, Tumor , Cell Survival/drug effects , Cell Survival/radiation effects , Coculture Techniques , Endothelial Cells/cytology , Endothelial Cells/metabolism , Gamma Rays , Immunohistochemistry , Mice , Microscopy, Fluorescence , Mitogen-Activated Protein Kinases/metabolism , Signal Transduction/drug effects , Signal Transduction/radiation effects , Spheroids, Cellular/cytology , Spheroids, Cellular/metabolism , Topotecan/toxicity
3.
J Control Release ; 217: 82-91, 2015 Nov 10.
Article in English | MEDLINE | ID: mdl-26310713

ABSTRACT

Reliable and predictive models of drug release kinetics in vitro and in vivo are still lacking for liposomal formulations. Developing robust, predictive release models requires systematic, quantitative characterization of these complex drug delivery systems with respect to the physicochemical properties governing the driving force for release. These models must also incorporate changes in release due to the dissolution media and methods employed to monitor release. This paper demonstrates the successful development and application of a mathematical mechanistic model capable of predicting doxorubicin (DXR) release kinetics from liposomal formulations resembling the FDA-approved nanoformulation DOXIL® using dynamic dialysis. The model accounts for DXR equilibria (e.g. self-association, precipitation, ionization), the change in intravesicular pH due to ammonia release, and dialysis membrane transport of DXR. The model was tested using a Box-Behnken experimental design in which release conditions including extravesicular pH, ammonia concentration in the release medium, and the dilution of the formulation (i.e. suspension concentration) were varied. Mechanistic model predictions agreed with observed DXR release up to 19h. The predictions were similar to a computer fit of the release data using an empirical model often employed for analyzing data generated from this type of experimental design. Unlike the empirical model, the mechanistic model was also able to provide reasonable predictions of release outside the tested design space. These results illustrate the usefulness of mechanistic modeling to predict drug release from liposomal formulations in vitro and its potential for future development of in vitro - in vivo correlations for complex nanoformulations.


Subject(s)
Antibiotics, Antineoplastic/chemistry , Doxorubicin/analogs & derivatives , Models, Chemical , Ammonia/chemistry , Doxorubicin/chemistry , Drug Liberation , Hydrogen-Ion Concentration , Membranes, Artificial , Polyethylene Glycols/chemistry , Renal Dialysis
4.
J Control Release ; 197: 10-9, 2015 Jan 10.
Article in English | MEDLINE | ID: mdl-25456833

ABSTRACT

A non-invasive fluorescence method was developed to monitor liposomal release kinetics of the anticancer agent topotecan (TPT) in physiological fluids and subsequently used to explore the cause of accelerated release in plasma. Analyses of fluorescence excitation spectra confirmed that unencapsulated TPT exhibits a red shift in its spectrum as pH is increased. This property was used to monitor TPT release from actively loaded liposomal formulations having a low intravesicular pH. Mathematical release models were developed to extract reliable rate constants for TPT release in aqueous solutions monitored by fluorescence and release kinetics obtained by HPLC. Using the fluorescence method, accelerated TPT release was observed in plasma as previously reported in the literature. Simulations to estimate the intravesicular pH were conducted to demonstrate that accelerated release correlated with alterations in the low intravesicular pH. This was attributed to the presence of ammonia in plasma samples rather than proteins and other plasma components generally believed to alter release kinetics in physiological samples. These findings shed light on the critical role that ammonia may play in contributing to the preclinical/clinical variability and performance seen with actively-loaded liposomal formulations of TPT and other weakly-basic anticancer agents.


Subject(s)
Topoisomerase I Inhibitors/blood , Topotecan/blood , Ammonia/chemistry , Humans , Liposomes , Models, Theoretical , Reproducibility of Results , Spectrometry, Fluorescence/methods , Topoisomerase I Inhibitors/chemistry , Topotecan/chemistry
5.
Pharm Res ; 32(5): 1752-63, 2015 May.
Article in English | MEDLINE | ID: mdl-25407546

ABSTRACT

PURPOSE: To better understand the mechanistic parameters that govern drug release from polymer micelles with acid-labile linkers. METHODS: A mathematical model was developed to describe drug release from block copolymer micelles composed of a poly(ethylene glycol) shell and a poly(aspartate) core, modified with drug binding linkers for pH-controlled release [hydrazide (HYD), aminobenzoate-hydrazide (ABZ), or glycine-hydrazide (GLY)]. Doxorubicin (Dox) was conjugated to the block copolymers through acid-labile hydrazone bonds. The polymer drug conjugates were used to prepare three polymer micelles (HYD-M, ABZ-M, and GLY-M). Drug release studies were performed to identify the factors governing pH-sensitive release of Dox. The effect of prolonged storage of copolymer material on release kinetics was also observed. RESULTS: Biphasic drug release kinetics were observed for all three micelle formulations. The developed model was able to quantify observed release kinetics upon the inclusion of terms for unconjugated Dox and two populations of conjugated Dox. Micelle/water partitioning of Dox was also incorporated into the model and found significant in all micelles under neutral conditions but reduced under acidic conditions. The drug binding linker played a major role in drug release as the extent of Dox release at specific time intervals was greater at pH 5.0 than at pH 7.4 (HYD-M > ABZ-M > GLY-M). Mathematical modeling was also able to correlate changes in release kinetics with the instability of the hydrazone conjugation of Dox during prolonged storage. CONCLUSION: These results illustrate the potential utility of mechanistic modeling to better assess release characteristics intrinsic to a particular drug/nanoparticle system.


Subject(s)
Antibiotics, Antineoplastic/administration & dosage , Delayed-Action Preparations/chemistry , Doxorubicin/administration & dosage , Hydrazones/chemistry , Micelles , Polymers/chemistry , Antibiotics, Antineoplastic/chemistry , Doxorubicin/chemistry , Drug Liberation , Drug Stability , Hydrogen-Ion Concentration , Kinetics , Models, Chemical
6.
Mol Pharm ; 11(4): 1314-25, 2014 Apr 07.
Article in English | MEDLINE | ID: mdl-24628304

ABSTRACT

Drug release from liposomal formulations is governed by a complex interplay of kinetic (i.e., drug permeability) and thermodynamic factors (i.e., drug partitioning to the bilayer surface). Release studies under sink conditions that attempt to mimic physiological conditions are insufficient to decipher these separate contributions. The present study explores release studies performed under nonsink conditions coupled with appropriate mathematical models to describe both the release kinetics and the conditions in which equilibrium is established. Liposomal release profiles for a model anticancer agent, topotecan, under nonsink conditions provided values for both the first-order rate constant for drug release and the bilayer/water partition coefficient. These findings were validated by conducting release studies under sink conditions via dynamic dialysis at the same temperature and buffer pH. A nearly identical rate constant for drug release could be obtained from dynamic dialysis data when appropriate volume corrections were applied and a mechanism-based mathematical model was employed to account for lipid bilayer binding and dialysis membrane transport. The usefulness of the nonsink method combined with mathematical modeling was further explored by demonstrating the effects of topotecan dimerization and bilayer surface charge potential on the bilayer/water partition coefficient at varying suspension concentrations of lipid and drug.


Subject(s)
Liposomes/metabolism , Topotecan/pharmacokinetics , Dimerization , Models, Theoretical , Permeability , Topotecan/chemistry
7.
J Control Release ; 174: 88-97, 2014 Jan 28.
Article in English | MEDLINE | ID: mdl-24231406

ABSTRACT

The use of liposomal delivery systems for the treatment of cancer has been extensively researched because of their passive targeting to the vasculature of solid tumors. While their potential to provide prolonged retention and high drug encapsulation is desirable for anticancer agents, a mechanistic understanding is required to optimize and design liposomal drug delivery systems capable of controllable release tailored to tumor type and patient. Topotecan (TPT) is a topoisomerase I inhibitor that undergoes reversible, pH-sensitive ring-opening hydrolysis. TPT may benefit from liposomal formulation using active loading strategies to generate low intravesicular pH to prolong drug retention and increase drug encapsulation. This paper develops a mathematical model to describe TPT's permeability as a function of pH by accounting for the drug's ionization state, membrane binding, and ring-opening interconversion kinetics. Studies were conducted to determine the acid dissociation constant of TPT's phenolic -OH and interconversion kinetics between TPT's lactone and carboxylate forms. Using the constants determined from these studies and release studies conducted at varying pH, permeability coefficients and membrane binding constants for each species of TPT were determined. Based on this model, three permeable species were observed. Interestingly, the two most permeable species were zwitterionic forms of TPT, and the permeability of the lactone zwitterion was comparable to that of the neutral form of another camptothecin analogue. Furthermore, release was affected by based-catalyzed interconversion kinetics between TPT's lactone and carboxylate forms. At neutral pH, release was rate-limited by formation of the TPT lactone from the ring-opened carboxylate form. Based on these findings, the developed model describing liposomal release of TPT may be used in the future to evaluate and optimize loading and subsequent release of liposomal TPT formulations utilizing active loading strategies.


Subject(s)
Liposomes/chemistry , Models, Biological , Topoisomerase I Inhibitors/chemistry , Topotecan/chemistry , Hydrogen-Ion Concentration , Hydrolysis , Kinetics , Membranes, Artificial , Phosphatidylcholines/chemistry , Phosphatidylethanolamines/chemistry , Polyethylene Glycols/chemistry
SELECTION OF CITATIONS
SEARCH DETAIL
...