Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 17 de 17
Filtrar
1.
J Control Release ; 366: 170-181, 2024 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-38128885

RESUMEN

The Port Delivery System with ranibizumab (PDS) is an innovative intraocular drug delivery system that has the potential to reduce treatment burden in patients with retinovascular diseases. The Port Delivery Platform (PD-P) implant is a permanent, indwelling device that can be refilled in situ through a self-sealing septum and is designed to continuously deliver ranibizumab by passive diffusion through a porous titanium release control element. We present results for the studies carried out to characterize the stability of ranibizumab for use with the PD-P. Simulated administration, in vitro release studies, and modeling studies were performed to evaluate the compatibility of ranibizumab with the PD-P administration components, and degradation and photostability in the implant. Simulated administration studies demonstrated that ranibizumab was highly compatible with the PD-P administration components (initial fill and refill needles) and commercially available administration components (syringe, transfer needle, syringe closure). Subsequent simulated in vitro release studies examining continuous delivery for up to 12 months in phosphate buffered saline, a surrogate for human vitreous, showed that the primary degradation products of ranibizumab were acidic variants. The presence of these variants increased over time and potency remained high. The stability attributes of ranibizumab were consistent across multiple implant refill-exchanges. Despite some degradation within the implant, the absolute mass of variants released daily from the implant was low due to the continuous release mechanism of the implant. Simulated light exposure within the implant resulted in small increases in the relative amount of ranibizumab degradants compared with those seen over 6 months.


Asunto(s)
Sistemas de Liberación de Medicamentos , Ranibizumab , Humanos , Difusión , Agujas , Porosidad
2.
Adv Funct Mater ; 23(29): 3628-3637, 2013 Aug 07.
Artículo en Inglés | MEDLINE | ID: mdl-25309305

RESUMEN

Superhydrophobic, porous, 3D materials composed of poly( ε -caprolactone) (PCL) and the hydrophobic polymer dopant poly(glycerol monostearate- co- ε -caprolactone) (PGC-C18) are fabricated using the electrospinning technique. These 3D materials are distinct from 2D superhydrophobic surfaces, with maintenance of air at the surface as well as within the bulk of the material. These superhydrophobic materials float in water, and when held underwater and pressed, an air bubble is released and will rise to the surface. By changing the PGC-C18 doping concentration in the meshes and/or the fiber size from the micro- to nanoscale, the long-term stability of the entrapped air layer is controlled. The rate of water infiltration into the meshes, and the resulting displacement of the entrapped air, is quantitatively measured using X-ray computed tomography. The properties of the meshes are further probed using surfactants and solvents of different surface tensions. Finally, the application of hydraulic pressure is used to quantify the breakthrough pressure to wet the meshes. The tools for fabrication and analysis of these superhydrophobic materials as well as the ability to control the robustness of the entrapped air layer are highly desirable for a number of existing and emerging applications.

3.
J Am Chem Soc ; 134(4): 2016-9, 2012 Feb 01.
Artículo en Inglés | MEDLINE | ID: mdl-22279966

RESUMEN

We have prepared 3D superhydrophobic materials from biocompatible building blocks, where air acts as a barrier component in a porous electrospun mesh to control the rate at which drug is released. Specifically, we fabricated poly(ε-caprolactone) electrospun meshes containing poly(glycerol monostearate-co-ε-caprolactone) as a hydrophobic polymer dopant, which results in meshes with a high apparent contact angle. We demonstrate that the apparent contact angle of these meshes dictates the rate at which water penetrates into the porous network and displaces entrapped air. The addition of a model bioactive agent (SN-38) showed a release rate with a striking dependence on the apparent contact angle that can be explained by this displacement of air within the electrospun meshes. We further show that porous electrospun meshes with higher surface area can be prepared that release more slowly than control nonporous constructs. Finally, the entrapped air layer within superhydrophobic meshes is shown to be robust in the presence of serum, as drug-loaded meshes were efficacious against cancer cells in vitro for >60 days, thus demonstrating their applicability for long-term drug delivery.


Asunto(s)
Materiales Biocompatibles/química , Portadores de Fármacos/química , Sistemas de Liberación de Medicamentos , Aire , Materiales Biocompatibles/síntesis química , Portadores de Fármacos/síntesis química , Interacciones Hidrofóbicas e Hidrofílicas , Tamaño de la Partícula , Poliésteres/química , Porosidad , Propiedades de Superficie
4.
Biomacromolecules ; 13(2): 406-11, 2012 Feb 13.
Artículo en Inglés | MEDLINE | ID: mdl-22242897

RESUMEN

A limitation to many polymer-based drug delivery systems is the lack of ability to customize a particular polymer composition for tailoring drug release kinetics to a specific clinical application. In this study, we investigated the structure-property effects of conjugating various hydrophobic biocompatible side chains to poly(glycerol-co-caprolactone) copolymers with the goal of achieving prolonged and controlled release of a chemotherapeutic agent. The choice of side chain significantly affected the resulting polymer properties including thermal transitions, relative crystallinity (ΔH(f)), and hydrophobicity. Drug-loaded films cast from solutions of polymer and 10-hydroxycamptothecin demonstrated prolonged release from four to over seven weeks depending upon side chain structure without initial burst release behavior. Use of the stearic acid-conjugated poly(glycerol-co-caprolactone) films afforded substantial anticancer activity in vitro for at least 50 days when exposed to fresh cultures of A549 human lung cancer cells over 24 h intervals, correlating well with the measured drug release kinetics.


Asunto(s)
Preparaciones de Acción Retardada/síntesis química , Portadores de Fármacos/síntesis química , Poliésteres/síntesis química , Antineoplásicos Fitogénicos/administración & dosificación , Antineoplásicos Fitogénicos/química , Antineoplásicos Fitogénicos/farmacología , Camptotecina/administración & dosificación , Camptotecina/análogos & derivados , Camptotecina/química , Camptotecina/farmacología , Línea Celular Tumoral , Supervivencia Celular/efectos de los fármacos , Preparaciones de Acción Retardada/química , Portadores de Fármacos/química , Humanos , Interacciones Hidrofóbicas e Hidrofílicas , Cinética , Neoplasias Pulmonares/tratamiento farmacológico , Neoplasias Pulmonares/patología , Espectroscopía de Resonancia Magnética , Tamaño de la Partícula , Poliésteres/química , Propiedades de Superficie
5.
J Control Release ; 345: 101-107, 2022 05.
Artículo en Inglés | MEDLINE | ID: mdl-35248647

RESUMEN

The Port Delivery System with ranibizumab (PDS) consists of an implant that is a permanent, indwelling drug delivery device that can be refilled through a self-sealing septum and is designed to continuously release a customized formulation of ranibizumab into the vitreous by passive diffusion through a porous titanium release control element. Target release rates of ranibizumab via the implant used in studies of the PDS in patients with neovascular age-related macular degeneration were selected based on clinical and pharmacokinetic (PK) data from previously conducted intravitreal ranibizumab injection studies. In-vitro testing was performed to verify release rates with a range of ranibizumab concentrations before the phase II Ladder (NCT02510794) and phase III Archway (NCT03677934) trials of the PDS. Implants were filled with ranibizumab and were regularly transferred to new buffer-containing tubes to represent ocular ranibizumab clearance and release kinetics. Ranibizumab concentrations were measured and release rates calculated. Release rate data were fit to an exponential model and compared with expected release kinetics of diffusion. Release profiles of the implant releasing ranibizumab at concentrations of 10 mg/mL, 40 mg/mL, and 100 mg/mL were determined in the pre-phase II in-vitro studies. At day 3.5, mean (SD) ranibizumab release rates were 1.75 (0.07), 6.42 (0.35), and 16.69 (0.67) µg/d for PDS 10 mg/mL, 40 mg/mL, and 100 mg/mL, respectively. At month 6, mean (SD) release rates were 1.68 (0.05) and 4.16 (0.05) µg/d for PDS 40 mg/mL and 100 mg/mL, respectively. Measured release rates were within 90% of theoretical release rates during the course of drug release. PDS 100 mg/mL released 73% (SD, 1.92) of drug by month 6. In the pre-phase III in-vitro studies, mean (SD) release rates with PDS 100 mg/mL were 17.97 (0.90), 4.44 (0.11), and 2.45 (0.08) µg/d at 3.5 days, 6 months, and 9 months, respectively. Cumulative release (SD) was 73% (1.92) by month 6 and 87% (1.88) by month 9. The sustained, continuous, and reproducible release from the PDS observed in the in-vitro studies was also observed in Ladder and Archway. In conclusion, in-vitro studies were a powerful tool for characterizing and verifying ranibizumab release from the PDS implant and supported clinical evaluation of the PDS. PDS 100 mg/mL, which was associated with the longest therapeutic-level delivery of ranibizumab among the concentrations tested, was selected for evaluation in the pivotal phase III Archway trial.


Asunto(s)
Sistemas de Liberación de Medicamentos , Liberación de Fármacos , Ranibizumab , Inhibidores de la Angiogénesis , Ensayos Clínicos Fase II como Asunto , Ensayos Clínicos Fase III como Asunto , Humanos , Inyecciones Intravítreas , Resultado del Tratamiento
6.
Biotechnol Bioeng ; 106(4): 627-37, 2010 Jul 01.
Artículo en Inglés | MEDLINE | ID: mdl-20229510

RESUMEN

Pre-filtration using ion exchange membrane adsorbers can improve parvovirus filter throughput of monoclonal antibodies (mAbs). The membranes work by binding trace foulants, and although some antibody product also binds, yields > or =99% are easily achieved by overloading. Results show that foulant adsorption is dependent on pH and conductivity, but independent of scale and adsorber brand. The ability to use ion exchange membranes as pre-filters is significant because it provides a clean, well defined, chemically stable option for enhancing throughput. Additionally, ion exchange membranes facilitate characterization of parvovirus filter foulants. Examination of adsorber elution samples using sedimentation velocity analysis and SEC-MALS/QELS revealed the presence of high molecular weight species ranging from 8 to 13 nm in hydrodynamic radius, which are similar in size to parvoviruses and thus would be expected to plug the pores of a parvovirus filter. A study of two identical membranes in-series supports the hypothesis that the foulants are soluble, trace level aggregates in the feed. This study's significance lies in a previously undiscovered application of membrane chromatography, leading to a more cost effective and robust approach to parvovirus filtration for the production of monoclonal antibodies.


Asunto(s)
Anticuerpos Monoclonales/biosíntesis , Anticuerpos Monoclonales/aislamiento & purificación , Filtración/métodos , Parvovirus/aislamiento & purificación , Medios de Cultivo/química , Conductividad Eléctrica , Concentración de Iones de Hidrógeno , Intercambio Iónico , Membranas
7.
Expert Opin Drug Deliv ; 16(1): 43-57, 2019 01.
Artículo en Inglés | MEDLINE | ID: mdl-30488721

RESUMEN

INTRODUCTION: Treating posterior eye diseases has become a major area of focus for pharmaceutical and biotechnology companies. Current standard of care for treating posterior eye diseases relies on administration via intravitreal injection. Although effective, this is not without complications and there is great incentive to develop longer-acting therapeutics and/or sustained release delivery systems. Here, we present an overview of emerging technologies for delivery of biologics to the back of the eye. AREAS COVERED: Posterior eye diseases, intravitreal injection, age-related macular degeneration, anti-VEGF, ocular pharmacokinetics, novel technologies to extend half-life, in vivo models, translation to the clinic, and hurdles to effective patient care. EXPERT OPINION: Posterior eye diseases are a worldwide public health issue. Although anti-VEGF molecules represent a major advance for treating diseases involving choroidal neovascularization, frequent injection can be burdensome for patients and clinicians. There is a need for effective and patient-friendly treatments for posterior eye diseases. Many technologies that enable long-acting delivery to the back of the eye are being evaluated. However, successful development of novel therapies and delivery technologies is hampered by a multitude of factors, including patient education, translatability of in vitro/in vivo preclinical data to the clinic, and regulatory challenges associated with novel technologies.


Asunto(s)
Sistemas de Liberación de Medicamentos , Oftalmopatías/tratamiento farmacológico , Degeneración Macular/tratamiento farmacológico , Animales , Disponibilidad Biológica , Ojo/metabolismo , Humanos , Inyecciones Intravítreas
8.
Biomaterials ; 29(13): 2033-48, 2008 May.
Artículo en Inglés | MEDLINE | ID: mdl-18289663

RESUMEN

Adsorption of lysozyme (Lys), human serum albumin (HSA), and immunoglobulin G (IgG) to anion- and cation-exchange resins is dominated by electrostatic interactions between protein and adsorbent. The solution-depletion method of measuring adsorption shows, however, that these proteins do not irreversibly adsorb to ion-exchange surfaces, even when the charge disparity between adsorbent and protein inferred from protein pI is large. Net-positively-charged Lys (pI=11) and net-negatively-charged HSA (pI=5.5) adsorb so strongly to sulfopropyl sepharose (SP; a negatively-charged, strong cation-exchange resin, -0.22 mmol/mL exchange capacity) that both resist displacement by net-neutral IgG (pI=7.0) in simultaneous adsorption competition experiments. By contrast, IgG readily displaces both Lys and HSA adsorbed either to quaternary ammonium sepharose (Q; a positively-charged, strong anion exchanger, +0.22 mmol/mL exchange capacity) or to octadecyl sepharose (ODS; a neutral hydrophobic resin, 0 mmol/mL exchange capacity). Thus it is concluded that adsorption results do not sensibly correlate with protein pI and that pI is actually a rather poor predictor of affinity for ion-exchange surfaces. Adsorption of Lys, HSA, and IgG to ion-exchange resins from stagnant solution leads to adsorbed multi-layers, into or onto which IgG adsorbs in adsorption competition experiments. Comparison of adsorption to ion-exchange resins and neutral ODS leads to the conclusion that the apparent standard free-energy of adsorption Delta Gads( degrees ) of Lys, HSA, and IgG is not large in comparison to thermal energy due to energy-compensating interactions between water, protein, and ion-exchange surfaces that leaves a small net Delta Gads( degrees ). Thus water is found to control protein adsorption to a full range of substratum types spanning hydrophobic (poorly water wettable) surfaces, hydrophilic surfaces bearing relatively-weak Lewis acid/base functionalities that wet with (hydrogen bond to) water but do not exhibit ion-exchange properties, and surfaces with strong Lewis acid/base functional groups that exhibit ion-exchange properties in the conventional chemistry sense of ion-exchange.


Asunto(s)
Inmunoglobulina G/química , Muramidasa/química , Albúmina Sérica/química , Adsorción , Humanos , Inmunoglobulina G/aislamiento & purificación , Iones/química , Muramidasa/aislamiento & purificación , Albúmina Sérica/aislamiento & purificación , Soluciones , Propiedades de Superficie
9.
J Control Release ; 273: 13-20, 2018 03 10.
Artículo en Inglés | MEDLINE | ID: mdl-29355621

RESUMEN

Monoclonal antibodies (mAbs), which are now more frequently administered by subcutaneous (SC) injection rather than intravenously, have become a tremendously successful drug format across a wide range of therapeutic areas. Preclinical evaluations of mAbs to be administered by SC injection are typically performed in species such as mice, rats, minipigs, and cynomolgus monkeys to obtain critical information regarding formulation performance and prediction of PK/PD outcomes needed to select clinical doses for first-in-human studies. Despite extensive efforts, no preclinical model has been identified to date that accurately predicts clinical outcomes for these SC injections. We have addressed this deficiency with a novel in vitro instrument, termed Scissor, to model events occurring at the SC injection site and now further validated this approach using a set of eight mAbs for which clinical PK/PD outcomes have been obtained. Diffusion of these mAbs from the Scissor system injection cartridge into a large volume physiological buffer, used to emulate mAb movement from the SC injection site into the systemic circulation, provided distinct profiles when monitored over a 6h period. Curve-fitting analysis of these profiles using the Hill equation identified parameters that were used, along with physicochemical properties for each mAb, in a partial least squares analysis to define a relationship between molecule and formulation properties with clinical PK outcomes. The results demonstrate that parameters of protein charge at neutral pH and isoelectric point (pI) along with combined formulation properties such as viscosity and mAb concentration can dictate the movement of the mAb from the injection cartridge to infinite sink compartment. Examination of profile characteristics of this movement provided a strong predictive correlation for these eight mAbs. Together, this approach demonstrates the feasibility of this in vitro modelling strategy as a tool to identify drug and formulation properties that can define the performance of SC injected medicines and provide the potential for predicting clinical outcomes that could be useful for formulation selection and a first-in-human clinical dosing strategy.


Asunto(s)
Anticuerpos Monoclonales/administración & dosificación , Anticuerpos Monoclonales/farmacocinética , Sistemas de Liberación de Medicamentos , Modelos Biológicos , Animales , Disponibilidad Biológica , Humanos , Inyecciones Subcutáneas , Macaca fascicularis , Ratones , Ratas , Porcinos , Porcinos Enanos
10.
Biomaterials ; 104: 87-103, 2016 10.
Artículo en Inglés | MEDLINE | ID: mdl-27449946

RESUMEN

Superhydrophobic surfaces are actively studied across a wide range of applications and industries, and are now finding increased use in the biomedical arena as substrates to control protein adsorption, cellular interaction, and bacterial growth, as well as platforms for drug delivery devices and for diagnostic tools. The commonality in the design of these materials is to create a stable or metastable air layer at the material surface, which lends itself to a number of unique properties. These activities are catalyzing the development of new materials, applications, and fabrication techniques, as well as collaborations across material science, chemistry, engineering, and medicine given the interdisciplinary nature of this work. The review begins with a discussion of superhydrophobicity, and then explores biomedical applications that are utilizing superhydrophobicity in depth including material selection characteristics, in vitro performance, and in vivo performance. General trends are offered for each application in addition to discussion of conflicting data in the literature, and the review concludes with the authors' future perspectives on the utility of superhydrophobic biomaterials for medical applications.


Asunto(s)
Materiales Biocompatibles/síntesis química , Materiales Biomiméticos/química , Interacciones Hidrofóbicas e Hidrofílicas , Nanocápsulas/química , Tensoactivos/química , Agua/química , Adsorción , Nanocápsulas/ultraestructura , Propiedades de Superficie
11.
Adv Healthc Mater ; 4(11): 1654-7, 2015 Aug 05.
Artículo en Inglés | MEDLINE | ID: mdl-26097150

RESUMEN

Rapid, simple, and inexpensive point-of-care (POC) medical tests are of significant need around the world. The transition between nonwetting and wetted states is used to create instrument-free surface tension sensors for POC diagnosis, using a layered electrospun mesh with incorporated dye to change color upon wetting.


Asunto(s)
Sistemas de Atención de Punto , Animales , Ácidos y Sales Biliares/orina , Glicerol/química , Leche/química , Poliésteres/química , Polímeros/química , Tensión Superficial , Tensoactivos , Humectabilidad
12.
J Control Release ; 214: 23-9, 2015 Sep 28.
Artículo en Inglés | MEDLINE | ID: mdl-26160309

RESUMEN

Layered superhydrophobic electrospun meshes composed of poly(ε-caprolactone) (PCL) and poly(glycerol monostearate-co-ε-caprolactone) (PGC-C18) are described as a local source of chemotherapeutic delivery. Specifically, the chemotherapeutic agent SN-38 is incorporated into a central 'core' layer, between two 'shield' layers of mesh without drug. This mesh is resistant to wetting of the surface and throughout the bulk due to the pronounced hydrophobicity imparted by the high roughness of a hydrophobic polymer, PGC-C18. In serum solution, these meshes exhibit slow initial drug release over 10days corresponding to media infiltrating the shield layer, followed by steady release over >30days, as the drug-loaded core layer is wetted. This sequence of events is supported by X-ray computed tomography imaging of a contrast agent solution infiltrating the mesh. In vitro cytotoxicity data collected with Lewis Lung Carcinoma (LLC) cells are consistent with this release profile, remaining cytotoxic for over 20days, longer than the unlayered version. Finally, after subcutaneous implantation in rats, histology of meshes with and without drug demonstrated good integration and lack of adverse reaction over 28days. The drug release rates, robust superhydrophobicity, in vitro cytotoxicity of SN-38 loaded meshes, and compatibility provide key design parameters for the development of an implantable chemotherapeutic-loaded device for the prevention of local lung cancer recurrence following surgical resection.


Asunto(s)
Preparaciones de Acción Retardada/química , Animales , Antineoplásicos Fitogénicos/administración & dosificación , Antineoplásicos Fitogénicos/uso terapéutico , Camptotecina/administración & dosificación , Camptotecina/análogos & derivados , Camptotecina/uso terapéutico , Carcinoma Pulmonar de Lewis/patología , Línea Celular Tumoral , Sistemas de Liberación de Medicamentos , Interacciones Hidrofóbicas e Hidrofílicas , Irinotecán , Recurrencia Local de Neoplasia/prevención & control , Poliésteres , Ratas , Ratas Endogámicas Lew , Solubilidad , Propiedades de Superficie
13.
J Control Release ; 214: 94-102, 2015 Sep 28.
Artículo en Inglés | MEDLINE | ID: mdl-26210441

RESUMEN

Subcutaneous (SC) injection is becoming a more common route for the administration of biopharmaceuticals. Currently, there is no reliable in vitro method that can be used to anticipate the in vivo performance of a biopharmaceutical formulation intended for SC injection. Nor is there an animal model that can predict in vivo outcomes such as bioavailability in humans. We address this unmet need by the development of a novel in vitro system, termed Scissor (Subcutaneous Injection Site Simulator). The system models environmental changes that a biopharmaceutical could experience as it transitions from conditions of a drug product formulation to the homeostatic state of the hypodermis following SC injection. Scissor uses a dialysis-based injection chamber, which can incorporate various concentrations and combinations of acellular extracellular matrix (ECM) components that may affect the release of a biopharmaceutical from the SC injection site. This chamber is immersed in a container of a bicarbonate-based physiological buffer that mimics the SC injection site and the infinite sink of the body. Such an arrangement allows for real-time monitoring of the biopharmaceutical within the injection chamber, and can be used to characterize physicochemical changes of the drug and its interactions with ECM components. Movement of a biopharmaceutical from the injection chamber to the infinite sink compartment simulates the drug migration from the injection site and uptake by the blood and/or lymph capillaries. Here, we present an initial evaluation of the Scissor system using the ECM element hyaluronic acid and test formulations of insulin and four different monoclonal antibodies. Our findings suggest that Scissor can provide a tractable method to examine the potential fate of a biopharmaceutical formulation after its SC injection in humans and that this approach may provide a reliable and representative alternative to animal testing for the initial screening of SC formulations.


Asunto(s)
Productos Biológicos/administración & dosificación , Productos Biológicos/farmacocinética , Anticuerpos Monoclonales/administración & dosificación , Anticuerpos Monoclonales/química , Anticuerpos Monoclonales/farmacocinética , Bicarbonatos/química , Productos Biológicos/química , Tampones (Química) , Química Farmacéutica , Diálisis , Diseño de Fármacos , Matriz Extracelular/metabolismo , Humanos , Ácido Hialurónico/metabolismo , Inyecciones Subcutáneas , Linfa/metabolismo , Tejido Subcutáneo/metabolismo
14.
Chem Commun (Camb) ; 49(8): 804-6, 2013 Jan 28.
Artículo en Inglés | MEDLINE | ID: mdl-23235806

RESUMEN

This work demonstrates a facile fabrication method to produce superhydrophobic coatings on chemically distinct materials using the electrospraying process. Coatings are mechanically robust, three-dimensional, and formed using a single fabrication step.


Asunto(s)
Glicerol/análogos & derivados , Poliésteres/química , Estearatos/química , Técnicas Electroquímicas , Glicerol/química , Interacciones Hidrofóbicas e Hidrofílicas , Monoglicéridos , Propiedades de Superficie
15.
Biol Direct ; 8: 7, 2013 Mar 22.
Artículo en Inglés | MEDLINE | ID: mdl-23522082

RESUMEN

BACKGROUND: The Michaelis-Menten equation, proposed a century ago, describes the kinetics of enzyme-catalyzed biochemical reactions. Since then, this equation has been used in countless, increasingly complex models of cellular metabolism, often including time-dependent enzyme levels. However, even for a single reaction, there remains a fundamental disconnect between our understanding of the reaction kinetics, and the regulation of that reaction through changes in the abundance of active enzyme. RESULTS: We revisit the Michaelis-Menten equation under the assumption of a time-dependent enzyme concentration. We show that all temporal enzyme profiles with the same average enzyme level yield identical substrate degradation- a simple analytical conclusion that can be thought of as an invariance principle, and which we validate experimentally using a ß-galactosidase assay. The ensemble of all time-dependent enzyme trajectories with the same average concentration constitutes a space of functions. We develop a simple model of biological fitness which assigns a cost to each of these trajectories (in the form of a function of functions, i.e. a functional). We then show how one can use variational calculus to analytically infer temporal enzyme profiles that minimize the overall enzyme cost. In particular, by separately treating the static costs of amino acid sequestration and the dynamic costs of protein production, we identify a fundamental cellular tradeoff. CONCLUSIONS: The overall metabolic outcome of a reaction described by Michaelis-Menten kinetics is ultimately determined by the average concentration of the enzyme during a given time interval. This invariance in analogy to path-independent phenomena in physics, suggests a new way in which variational calculus can be employed to address biological questions. Together, our results point to possible avenues for a unified approach to studying metabolism and its regulation. REVIEWERS: This article was reviewed by Sergei Maslov, William Hlavacek and Daniel Kahn.


Asunto(s)
Enzimas/metabolismo , Cinética
16.
Adv Healthc Mater ; 2(9): 1204-8, 2013 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-23592698

RESUMEN

Application of high-intensity focused ultrasound to drug-loaded superhydrophobic meshes affords triggered drug release by displacing an entrapped air layer. The air layer within the superhydrophobic meshes is characterized using direct visualization and B-mode imaging. Drug-loaded superhydrophobic meshes are cytotoxic in an in vitro assay after ultrasound treatment.


Asunto(s)
Portadores de Fármacos/química , Ultrasonido , Antineoplásicos Fitogénicos/química , Antineoplásicos Fitogénicos/toxicidad , Camptotecina/análogos & derivados , Camptotecina/química , Camptotecina/toxicidad , Supervivencia Celular/efectos de los fármacos , Humanos , Interacciones Hidrofóbicas e Hidrofílicas , Irinotecán , Células MCF-7 , Poliésteres/química
17.
J Control Release ; 162(1): 92-101, 2012 Aug 20.
Artículo en Inglés | MEDLINE | ID: mdl-22684120

RESUMEN

In this work we expand upon a recently reported local drug delivery device, where air is used as a degradable component of our material to control drug release (J. Am. Chem. Soc. 2012, 134, 2016-2019). We consider its potential use as a drug loaded strip to provide both mechanical stability to the anastomosis, and as a means to release drug locally over prolonged periods for prevention of locoregional recurrence in colorectal cancer. Specifically, we electrospun poly(ε-caprolactone) (PCL) with the hydrophobic polymer dopant poly(glycerol monostearate-co-ε-caprolactone) (PGC-C18) and used the resultant mesh to control the release of two anticancer drugs (CPT-11 and SN-38). The increase in mesh hydrophobicity with PGC-C18 addition slows drug release both by the traditional means of drug diffusion, as well as by increasing the stability of the entrapped air layer to delay drug release. We demonstrate that superhydrophobic meshes have mechanical properties appropriate for surgical buttressing of the anastomosis, permit non-invasive assessment of mesh location and documentation of drug release via ultrasound, and release chemotherapy over a prolonged period of time (>90 days) resulting in significant tumor cytotoxicity against a human colorectal cell line (HT-29).


Asunto(s)
Antineoplásicos Fitogénicos/administración & dosificación , Camptotecina/análogos & derivados , Neoplasias Colorrectales/tratamiento farmacológico , Sistemas de Liberación de Medicamentos/instrumentación , Glicerol/análogos & derivados , Poliésteres/química , Estearatos/química , Antineoplásicos Fitogénicos/farmacología , Camptotecina/administración & dosificación , Camptotecina/farmacología , Línea Celular Tumoral , Supervivencia Celular/efectos de los fármacos , Glicerol/química , Humanos , Interacciones Hidrofóbicas e Hidrofílicas , Irinotecán , Monoglicéridos
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA