Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 19 de 19
Filter
Add more filters










Publication year range
1.
Mol Pharm ; 21(7): 3310-3320, 2024 Jul 01.
Article in English | MEDLINE | ID: mdl-38856116

ABSTRACT

Siponimod is a promising agent for the inhibition of ocular neovascularization in diabetic retinopathy and age-related macular degeneration. Siponimod's development for ophthalmological application is hindered by the limited information available on the drug's solubility, stability, ocular pharmacokinetics (PK), and toxicity in vivo. In this study, we investigated the aqueous stability of siponimod under stress conditions (up to 60 °C) and its degradation behavior in solution. Additionally, siponimod's ocular PK and toxicity were investigated using intravitreal injection of two different doses (either 1300 or 6500 ng) in an albino rabbit model. Siponimod concentration was quantified in the extracted vitreous, and the PK parameters were calculated. The drug half-life after administration of the low and high doses was 2.8 and 3.9 h, respectively. The data obtained in vivo was used to test the ability of published in silico models to predict siponimod's PK accurately. Two models that correlated siponimod's molecular descriptors with its elimination from the vitreous closely predicted the half-life. Furthermore, 24 h and 7 days after intravitreal injections, the retinas showed no signs of toxicity. This study provides important information necessary for the formulation and development of siponimod for ophthalmologic applications. The short half-life of siponimod necessitates the development of a sustained drug delivery system to maintain therapeutic concentrations over an extended period, while the lack of short-term ocular toxicity observed in the retinas of siponimod-treated rabbits supports possible clinical use.


Subject(s)
Azetidines , Intravitreal Injections , Animals , Rabbits , Azetidines/pharmacokinetics , Azetidines/administration & dosage , Half-Life , Vitreous Body/drug effects , Vitreous Body/metabolism , Male , Retina/drug effects , Retina/metabolism , Eye/drug effects , Eye/metabolism , Diabetic Retinopathy/drug therapy , Angiogenesis Inhibitors/pharmacokinetics , Angiogenesis Inhibitors/administration & dosage , Angiogenesis Inhibitors/toxicity , Solubility , Macular Degeneration/drug therapy , Benzyl Compounds
2.
J Control Release ; 366: 812-833, 2024 Feb.
Article in English | MEDLINE | ID: mdl-38101753

ABSTRACT

In the past decade RNA-based therapies such as small interfering RNA (siRNA) and messenger RNA (mRNA) have emerged as new and ground-breaking therapeutic agents for the treatment and prevention of many conditions from viral infection to cancer. Most clinically approved RNA therapies are parenterally administered which impacts patient compliance and adds to healthcare costs. Pulmonary administration via inhalation is a non-invasive means to deliver RNA and offers an attractive alternative to injection. Nebulisation is a particularly appealing method due to the capacity to deliver large RNA doses during tidal breathing. In this review, we discuss the unique physiological barriers presented by the lung to efficient nebulised RNA delivery and approaches adopted to circumvent this problem. Additionally, the different types of nebulisers are evaluated from the perspective of their suitability for RNA delivery. Furthermore, we discuss recent preclinical studies involving nebulisation of RNA and analysis in in vitro and in vivo settings. Several studies have also demonstrated the importance of an effective delivery vector in RNA nebulisation therefore we assess the variety of lipid, polymeric and hybrid-based delivery systems utilised to date. We also consider the outlook for nebulised RNA medicinal products and the hurdles which must be overcome for successful clinical translation. In summary, nebulised RNA delivery has demonstrated promising potential for the treatment of several lung-related conditions such as asthma, COPD and cystic fibrosis, to which the mode of delivery is of crucial importance for clinical success.


Subject(s)
Asthma , Respiratory Aerosols and Droplets , Humans , Cytosol , RNA, Small Interfering , Lung
4.
J Pharmacol Exp Ther ; 386(2): 224-241, 2023 08.
Article in English | MEDLINE | ID: mdl-37188532

ABSTRACT

Sphingosine-1-phosphate (S1P) receptors control endothelial cell proliferation, migration, and survival. Evidence of the ability of S1P receptor modulators to influence multiple endothelial cell functions suggests their potential use for antiangiogenic effect. The main purpose of our study was to investigate the potential of siponimod for the inhibition of ocular angiogenesis in vitro and in vivo. We investigated the effects of siponimod on the metabolic activity (thiazolyl blue tetrazolium bromide assay), cell toxicity (lactate dehydrogenase release), basal proliferation and growth factor-induced proliferation (bromodeoxyuridine assay), and migration (transwell migration assay) of human umbilical vein endothelial cells (HUVEC) and retinal microvascular endothelial cells (HRMEC). The effects of siponimod on HRMEC monolayer integrity, barrier function under basal conditions, and tumor necrosis factor alpha (TNF-α)-induced disruption were assessed using the transendothelial electrical resistance and fluorescein isothiocyanate-dextran permeability assays. Siponimod's effect on TNF-α-induced distribution of barrier proteins in HRMEC was investigated using immunofluorescence. Finally, the effect of siponimod on ocular neovascularization in vivo was assessed using suture-induced corneal neovascularization in albino rabbits. Our results show that siponimod did not affect endothelial cell proliferation or metabolic activity but significantly inhibited endothelial cell migration, increased HRMEC barrier integrity, and reduced TNF-α-induced barrier disruption. Siponimod also protected against TNF-α-induced disruption of claudin-5, zonula occludens-1, and vascular endothelial-cadherin in HRMEC. These actions are mainly mediated by sphingosine-1-phosphate receptor 1 modulation. Finally, siponimod prevented the progression of suture-induced corneal neovascularization in albino rabbits. In conclusion, the effects of siponimod on various processes known to be involved in angiogenesis support its therapeutic potential in disorders associated with ocular neovascularization. SIGNIFICANCE STATEMENT: Siponimod is an extensively characterized sphingosine-1-phosphate receptor modulator already approved for the treatment of multiple sclerosis. It inhibited retinal endothelial cell migration, potentiated endothelial barrier function, protected against tumor necrosis factor alpha-induced barrier disruption, and also inhibited suture-induced corneal neovascularization in rabbits. These results support its use for a novel therapeutic indication in the management of ocular neovascular diseases.


Subject(s)
Corneal Neovascularization , Tumor Necrosis Factor-alpha , Animals , Humans , Rabbits , Retina , Neovascularization, Pathologic , Human Umbilical Vein Endothelial Cells , Cells, Cultured
5.
Pharmaceutics ; 15(1)2023 Jan 11.
Article in English | MEDLINE | ID: mdl-36678871

ABSTRACT

(1) Background: Three-dimensional (3D) in vitro, biorelevant culture models that recapitulate cancer progression can help elucidate physio-pathological disease cues and enhance the screening of more effective therapies. Insufficient research has been conducted to generate in vitro 3D models to replicate the spread of prostate cancer to the bone, a key metastatic site of the disease, and to understand the interplay between the key cell players. In this study, we aim to investigate PLGA and nano-hydroxyapatite (nHA)/PLGA mixed scaffolds as a predictive preclinical tool to study metastatic prostate cancer (mPC) in the bone and reduce the gap that exists with traditional 2D cultures. (2) Methods: nHA/PLGA mixed scaffolds were produced by electrospraying, compacting, and foaming PLGA polymer microparticles, +/- nano-hydroxyapatite (nHA), and a salt porogen to produce 3D, porous scaffolds. Physicochemical scaffold characterisation together with an evaluation of osteoblastic (hFOB 1.19) and mPC (PC-3) cell behaviour (RT-qPCR, viability, and differentiation) in mono- and co-culture, was undertaken. (3) Results: The results show that the addition of nHA, particularly at the higher-level impacted scaffolds in terms of mechanical and degradation behaviour. The nHA 4 mg resulted in weaker scaffolds, but cell viability increased. Qualitatively, fluorescent imaging of cultures showed an increase in PC-3 cells compared to osteoblasts despite lower initial PC-3 seeding densities. Osteoblast monocultures, in general, caused an upregulation (or at least equivalent to controls) in gene production, which was highest in plain scaffolds and decreased with increases in nHA. Additionally, the genes were downregulated in PC3 and co-cultures. Further, drug toxicity tests demonstrated a significant effect in 2D and 3D co-cultures. (4) Conclusions: The results demonstrate that culture conditions and environment (2D versus 3D, monoculture versus co-culture) and scaffold composition all impact cell behaviour and model development.

6.
Drug Deliv Transl Res ; 13(8): 2110-2141, 2023 08.
Article in English | MEDLINE | ID: mdl-35930221

ABSTRACT

Acute myeloid leukemia (AML) is a heterogeneous malignancy affecting myeloid cells in the bone marrow (BM) but can spread giving rise to impaired hematopoiesis. AML incidence increases with age and is associated with poor prognostic outcomes. There has been a disconnect between the success of novel drug compounds observed in preclinical studies of hematological malignancy and less than exceptional therapeutic responses in clinical trials. This review aims to provide a state-of-the-art overview on the different preclinical models of AML available to expand insights into disease pathology and as preclinical screening tools. Deciphering the complex physiological and pathological processes and developing predictive preclinical models are key to understanding disease progression and fundamental in the development and testing of new effective drug treatments. Standard scaffold-free suspension models fail to recapitulate the complex environment where AML occurs. To this end, we review advances in scaffold/matrix-based 3D models and outline the most recent advances in on-chip technology. We also provide an overview of clinically relevant animal models and review the expanding use of patient-derived samples, which offer the prospect to create more "patient specific" screening tools either in the guise of 3D matrix models, microphysiological "organ-on-chip" tools or xenograft models and discuss representative examples.


Subject(s)
Leukemia, Myeloid, Acute , Animals , Humans , Leukemia, Myeloid, Acute/drug therapy , Leukemia, Myeloid, Acute/pathology , Bone Marrow/pathology , Disease Models, Animal
7.
Antibiotics (Basel) ; 11(12)2022 Dec 15.
Article in English | MEDLINE | ID: mdl-36551479

ABSTRACT

Orthopaedic device implants play a crucial role in restoring functionality to patients suffering from debilitating musculoskeletal diseases or to those who have experienced traumatic injury. However, the surgical implantation of these devices carries a risk of infection, which represents a significant burden for patients and healthcare providers. This review delineates the pathogenesis of orthopaedic implant infections and the challenges that arise due to biofilm formation and the implications for treatment. It focuses on research advancements in the development of next-generation orthopaedic medical devices to mitigate against implant-related infections. Key considerations impacting the development of devices, which must often perform multiple biological and mechanical roles, are delineated. We review technologies designed to exert spatial and temporal control over antimicrobial presentation and the use of antimicrobial surfaces with intrinsic antibacterial activity. A range of measures to control bio-interfacial interactions including approaches that modify implant surface chemistry or topography to reduce the capacity of bacteria to colonise the surface, form biofilms and cause infections at the device interface and surrounding tissues are also reviewed.

8.
Adv Drug Deliv Rev ; 187: 114342, 2022 08.
Article in English | MEDLINE | ID: mdl-35569559

ABSTRACT

There is an increasing momentum in research and pharmaceutical industry communities to design sustained, non-invasive delivery systems to treat chronic neovascular ocular diseases that affect the posterior segment of the eye including age-related macular degeneration and diabetic retinopathy. Current treatments include VEGF blockers, which have revolutionized the standard of care for patients, but their maximum therapeutic benefit is hampered by the need for recurrent and invasive administration procedures. Currently approved delivery systems intended to address these limitations exploit polymer technology to regulate drug release in a sustained manner. Here, we critically review sustained drug delivery approaches for the treatment of chronic neovascular diseases affecting the ocular posterior segment, with a special emphasis on novel and polymeric technologies spanning the spectrum of preclinical and clinical investigation, and those approved for treatment. The mechanism by which each formulation imparts sustained release, the impact of formulation characteristics on release and foreign body reaction, and special considerations related to the translation of these systems are discussed.


Subject(s)
Diabetic Retinopathy , Macular Degeneration , Diabetic Retinopathy/drug therapy , Drug Delivery Systems/methods , Eye , Humans , Macular Degeneration/drug therapy , Polymers/therapeutic use
9.
Br J Ophthalmol ; 106(9): 1187-1195, 2022 09.
Article in English | MEDLINE | ID: mdl-33962970

ABSTRACT

Neovascular ocular diseases (such as age-related macular degeneration, diabetic retinopathy and retinal vein occlusion) are characterised by common pathological processes that contribute to disease progression. These include angiogenesis, oedema, inflammation, cell death and fibrosis. Currently available therapies target the effects of vascular endothelial growth factor (VEGF), the main mediator of pathological angiogenesis. Unfortunately, VEGF blockers are expensive biological therapeutics that necessitate frequent intravitreal administration and are associated with multiple adverse effects. Thus, alternative treatment options associated with fewer side effects are required for disease management. This review introduces sphingosine 1-phosphate (S1P) as a potential pharmacological target for the treatment of neovascular ocular pathologies. S1P is a sphingolipid mediator that controls cellular growth, differentiation, survival and death. S1P actions are mediated by five G protein-coupled receptors (S1P1-5 receptors) which are abundantly expressed in all retinal and subretinal structures. The action of S1P on S1P1 receptors can reduce angiogenesis, increase endothelium integrity, reduce photoreceptor apoptosis and protect the retina against neurodegeneration. Conversely, S1P2 receptor signalling can increase neovascularisation, disrupt endothelial junctions, stimulate VEGF release, and induce retinal cell apoptosis and degeneration of neural retina. The aim of this review is to thoroughly discuss the role of S1P and its different receptor subtypes in angiogenesis, inflammation, apoptosis and fibrosis in order to determine which of these S1P-mediated processes may be targeted therapeutically.


Subject(s)
Diabetic Retinopathy , Vascular Endothelial Growth Factor A , Fibrosis , Humans , Inflammation , Lysophospholipids , Neovascularization, Pathologic , Sphingosine/analogs & derivatives , Vascular Endothelial Growth Factors
10.
Drug Deliv Transl Res ; 11(2): 702-716, 2021 04.
Article in English | MEDLINE | ID: mdl-33713316

ABSTRACT

Poor integration of orthopaedic devices with the host tissue owing to aseptic loosening and device-associated infections are two of the leading causes of implant failure, which represents a significant problem for both patients and the healthcare system. Novel strategies have focused on silver to combat antimicrobial infections as an alternative to drug therapeutics. In this study, we investigated the impact of increasing the % substitution (12% wt) of silver and strontium in hydroxyapatite (HA) coatings to enhance antimicrobial properties and stimulate osteoblasts, respectively. Additionally, we prepared a binary substituted coating containing both silver and strontium (AgSrA) at 12% wt as a comparison. All coatings were deposited using a novel blasting process, CoBlast, onto biomedical grade titanium (V). Surface physicochemical properties, cytocompatibility and antimicrobial functionality were determined. The anticolonising properties of the coatings were screened using Staphylococcus aureus ATCC 1448, and thereafter, the AgA coating was evaluated using clinically relevant strains. Strontium-doped surfaces demonstrated enhanced osteoblast viability; however, a lower inhibition of biofilm formation was observed compared with the other surfaces. A co-substituted AgSrA surface did not show enhanced osteoblast or anticolonising properties compared with the SrA and AgA surfaces, respectively. Due to its superior anticolonising performance in preliminary studies, AgA was chosen for further studies. The AgA coated surfaces demonstrated good antibacterial activity (eluted and immobilised ion) against methicillin-resistant S. aureus followed by methicillin-sensitive Staphylococcus aureus clinical isolates; however, the AgA surface displayed poor impact against Staphylococcus epidermidis. In conclusion, herein, we demonstrate that HA can be substituted with a range of ions to augment the properties of HA coatings on orthopaedic devices, which offer promising potential to combat orthopaedic device-associated infections and enhance device performance.


Subject(s)
Methicillin-Resistant Staphylococcus aureus , Orthopedics , Anti-Bacterial Agents/pharmacology , Biofilms , Coated Materials, Biocompatible , Durapatite , Humans , Silver/pharmacology , Strontium , Surface Properties , Titanium
11.
Int J Pharm ; 590: 119956, 2020 Nov 30.
Article in English | MEDLINE | ID: mdl-33035608

ABSTRACT

Sphingosine 1-phosphate (S1P) receptor modulators can influence bone regeneration owing to their positive impact on osteoblast differentiation and neovascularisation. While previous studies have utilised non-specific S1P and fingolimod, this study aims to design and characterise a controlled release vehicle to deliver the specific S1P1 & 5 receptor modulator siponimod and test its effectiveness in rat critical cranial defects. Electrospun scaffolds of poly lactide-co-glycolide (PLGA) were loaded with siponimod at drug:polymer mass ratios of 0.5:100 to 2:100. Where indicated, collagen was co-spun at a collagen:polymer mass ratio of 2:100. Thereafter, scaffolds underwent in vitro physicochemical characterisation and in vivo assessment using a rat cranial defect model. Drug-loaded scaffolds showed controlled release of siponimod, -cytocompatibility with endothelial and osteoblast cells in vitro, and furthermore, showed that released siponimod stimulated osteoblast differentiation and endothelial cell migration. The in vivo cranial defect repair study showed regeneration was occurring in the defect, although there was no significant difference in the extent of mineralisation between scaffold experimental groups. To our knowledge, this is the first study investigating siponimod in bone regeneration. In vitro studies confirm a positive impact on key cells involved in bone regeneration, however, the scaffolds did not result in significant repair of critical cranial defects.


Subject(s)
Polymers , Tissue Scaffolds , Animals , Azetidines , Benzyl Compounds , Bone Regeneration , Rats
12.
Eur J Pharmacol ; 882: 173262, 2020 Sep 05.
Article in English | MEDLINE | ID: mdl-32534075

ABSTRACT

The repair of critical bone defects remains a significant therapeutic challenge. While the implantation of drug-eluting scaffolds is an option, a drug with the optimal pharmacological properties has not yet been identified. Agents acting at sphingosine 1-phosphate (S1P) receptors have been considered, but those investigated so far do not discriminate between the five known S1P receptors. This work was undertaken to investigate the potential of the specific S1P1/5 modulator siponimod as a bone regenerative agent, by testing in vitro its effect on cell types critical to the bone regeneration process. hFOB osteoblasts and HUVEC endothelial cells were treated with siponimod and other S1P receptor modulators and investigated for changes in intracellular cyclic AMP content, viability, proliferation, differentiation, attachment and cellular motility. Siponimod showed no effect on the viability and proliferation of osteoblasts and endothelial cells, but increased osteoblast differentiation (as shown by increased alkaline phosphatase activity). Furthermore, siponimod significantly increased endothelial cell motility in scratch and transwell migration assays. These effects on osteoblast differentiation and endothelial cell migration suggest that siponimod may be a potential agent for the stimulation of localised differentiation of osteoblasts in critical bone defects.


Subject(s)
Azetidines/pharmacology , Benzyl Compounds/pharmacology , Bone Regeneration/drug effects , Human Umbilical Vein Endothelial Cells/drug effects , Osteoblasts/drug effects , Sphingosine 1 Phosphate Receptor Modulators/pharmacology , Cell Physiological Phenomena/drug effects , Cells, Cultured , Cyclic AMP/metabolism , Fingolimod Hydrochloride/pharmacology , Human Umbilical Vein Endothelial Cells/physiology , Humans , Osteoblasts/physiology
13.
Pharmacol Res ; 125(Pt B): 232-245, 2017 Nov.
Article in English | MEDLINE | ID: mdl-28855094

ABSTRACT

The lipid mediator sphingosine 1-phosphate (S1P) affects cellular functions in most systems. Interest in its therapeutic potential has increased following the discovery of its G protein-coupled receptors and the recent availability of agents that can be safely administered in humans. Although the role of S1P in bone biology has been the focus of much less research than its role in the nervous, cardiovascular and immune systems, it is becoming clear that this lipid influences many of the functions, pathways and cell types that play a key role in bone maintenance and repair. Indeed, S1P is implicated in many osteogenesis-related processes including stem cell recruitment and subsequent differentiation, differentiation and survival of osteoblasts, and coupling of the latter cell type with osteoclasts. In addition, S1P's role in promoting angiogenesis is well-established. The pleiotropic effects of S1P on bone and blood vessels have significant potential therapeutic implications, as current therapeutic approaches for critical bone defects show significant limitations. Because of the complex effects of S1P on bone, the pharmacology of S1P-like agents and their physico-chemical properties, it is likely that therapeutic delivery of S1P agents will offer significant advantages compared to larger molecular weight factors. Hence, it is important to explore novel methods of utilizing S1P agents therapeutically, and improve our understanding of how S1P and its receptors modulate bone physiology and repair.


Subject(s)
Bone and Bones/metabolism , Lysophospholipids/metabolism , Osteogenesis/physiology , Sphingosine/analogs & derivatives , Animals , Humans , Neovascularization, Physiologic/physiology , Osteoblasts/metabolism , Osteoclasts/metabolism , Signal Transduction , Sphingosine/metabolism , Stem Cells/metabolism
14.
J Control Release ; 250: 86-95, 2017 03 28.
Article in English | MEDLINE | ID: mdl-28132935

ABSTRACT

Drug release from mesoporous silica systems has been widely investigated in vitro using USP Type II (paddle) dissolution apparatus. However, it is not clear if the observed enhanced in vitro dissolution can forecast drug bioavailability in vivo. In this study, the ability of different in vitro dissolution models to predict in vivo oral bioavailability in a pig model was examined. The fenofibrate-loaded mesoporous silica formulation was compared directly to a commercial reference product, Lipantil Supra®. Three in vitro dissolution methods were considered; USP Type II (paddle) apparatus, USP Type IV (flow-through cell) apparatus and a USP IV Transfer model (incorporating a SGF to FaSSIF-V2 media transfer). In silico modelling, using a physiologically based pharmacokinetic modelling and simulation software package (Gastroplus™), to generate in vitro/in vivo relationships, was also investigated. The study demonstrates that the in vitro dissolution performance of a mesoporous silica formulation varies depending on the dissolution apparatus utilised and experimental design. The findings show that the USP IV transfer model was the best predictor of in vivo bioavailability. The USP Type II (paddle) apparatus was not effective at forecasting in vivo behaviour. This observation is likely due to hydrodynamic differences between the two apparatus and the ability of the transfer model to better simulate gastrointestinal transit. The transfer model is advantageous in forecasting in vivo behaviour for formulations which promote drug supersaturation and as a result are prone to precipitation to a more energetically favourable, less soluble form. The USP IV transfer model could prove useful in future mesoporous silica formulation development. In silico modelling has the potential to assist in this process. However, further investigation is required to overcome the limitations of the model for solubility enhancing formulations.


Subject(s)
Drug Carriers/chemistry , Silicon Dioxide/chemistry , Animals , Biological Availability , Chemistry, Pharmaceutical , Computer Simulation , Drug Liberation , Female , Fenofibrate/chemistry , Fenofibrate/pharmacokinetics , Humans , Hydrodynamics , Models, Biological , Porosity , Solubility , Swine
15.
Expert Opin Drug Deliv ; 13(1): 93-108, 2016.
Article in English | MEDLINE | ID: mdl-26549623

ABSTRACT

INTRODUCTION: Silica materials, in particular mesoporous silicas, have demonstrated excellent properties to enhance the oral bioavailability of poorly water-soluble drugs. Current research in this area is focused on investigating the kinetic profile of drug release from these carriers and manufacturing approaches to scale-up production for commercial manufacture. AREAS COVERED: This review provides an overview of different methods utilized to load drugs onto mesoporous silica carriers. The influence of silica properties and silica pore architecture on drug loading and release are discussed. The kinetics of drug release from mesoporous silica systems is examined and the manufacturability and stability of these formulations are reviewed. Finally, the future prospects of mesoporous silica drug delivery systems are considered. EXPERT OPINION: Substantial progress has been made in the characterization and development of mesoporous drug delivery systems for drug dissolution enhancement. However, more research is required to fully understand the drug release kinetic profile from mesoporous silica materials. Incomplete drug release from the carrier and the possibility of drug re-adsorption onto the silica surface need to be investigated. Issues to be addressed include the manufacturability and regulation status of formulation approaches employing mesoporous silica to enhance drug dissolution. While more research is needed to support the move of this technology from the bench to a commercial medicinal product, it is a realistic prospect for the near future.


Subject(s)
Chemistry, Pharmaceutical/methods , Drug Carriers/pharmacokinetics , Drug Liberation , Silicon Dioxide/chemistry , Silicon Dioxide/pharmacokinetics , Biological Availability , Humans , Particle Size , Porosity , Solubility , Water
16.
Eur J Pharm Sci ; 50(3-4): 400-9, 2013 Nov 20.
Article in English | MEDLINE | ID: mdl-23981335

ABSTRACT

Loading a poorly water-soluble drug onto a high surface area carrier such as mesoporous silica (SBA-15) can increase the drug's dissolution rate and oral bioavailability. The loading method can influence subsequent drug properties including solid state structure and release rate. The objective of this research was to compare several loading processes in terms of drug distribution throughout the mesoporous silica matrix, drug solid state form and drug release properties. A model poorly water-soluble drug fenofibrate was loaded onto SBA-15 using; (i) physical mixing, (ii) melt, (iii) solvent impregnation, (iv) liquid CO2 and (v) supercritical CO2 methods. Physical mixing resulted in heterogeneous drug-loading, with no evidence of drug in the mesopores and the retention of the drug in its crystalline state. The other loading processes yielded more homogeneous drug-loading; the drug was deposited into the mesopores of the SBA-15 and was non-crystalline. All the processing methods resulted in enhanced drug release compared to the unprocessed drug with the impregnation, liquid and SC-CO2 producing the greatest increase at t=30 min.


Subject(s)
Fenofibrate/chemistry , Silicon Dioxide/chemistry , Drug Compounding/methods , Drug Delivery Systems , Drug Stability , Porosity
17.
J Nanobiotechnology ; 11: 1, 2013 Jan 23.
Article in English | MEDLINE | ID: mdl-23343139

ABSTRACT

BACKGROUND: Aerosolized therapeutics hold great potential for effective treatment of various diseases including lung cancer. In this context, there is an urgent need to develop novel nanocarriers suitable for drug delivery by nebulization. To address this need, we synthesized and characterized a biocompatible drug delivery vehicle following surface coating of Fe3O4 magnetic nanoparticles (MNPs) with a polymer poly(lactic-co-glycolic acid) (PLGA). The polymeric shell of these engineered nanoparticles was loaded with a potential anti-cancer drug quercetin and their suitability for targeting lung cancer cells via nebulization was evaluated. RESULTS: Average particle size of the developed MNPs and PLGA-MNPs as measured by electron microscopy was 9.6 and 53.2 nm, whereas their hydrodynamic swelling as determined using dynamic light scattering was 54.3 nm and 293.4 nm respectively. Utilizing a series of standardized biological tests incorporating a cell-based automated image acquisition and analysis procedure in combination with real-time impedance sensing, we confirmed that the developed MNP-based nanocarrier system was biocompatible, as no cytotoxicity was observed when up to 100 µg/ml PLGA-MNP was applied to the cultured human lung epithelial cells. Moreover, the PLGA-MNP preparation was well-tolerated in vivo in mice when applied intranasally as measured by glutathione and IL-6 secretion assays after 1, 4, or 7 days post-treatment. To imitate aerosol formation for drug delivery to the lungs, we applied quercitin loaded PLGA-MNPs to the human lung carcinoma cell line A549 following a single round of nebulization. The drug-loaded PLGA-MNPs significantly reduced the number of viable A549 cells, which was comparable when applied either by nebulization or by direct pipetting. CONCLUSION: We have developed a magnetic core-shell nanoparticle-based nanocarrier system and evaluated the feasibility of its drug delivery capability via aerosol administration. This study has implications for targeted delivery of therapeutics and poorly soluble medicinal compounds via inhalation route.


Subject(s)
Drug Delivery Systems/methods , Magnetics , Nanoparticles/chemistry , Quercetin/pharmacology , Aerosols , Animals , Antineoplastic Agents/pharmacology , Cell Line, Tumor , Female , Glutathione/analysis , Humans , Interleukin-6/analysis , Lactic Acid/chemistry , Mice , Mice, Inbred BALB C , Microscopy, Electron, Transmission , Particle Size , Polyglycolic Acid/chemistry , Polylactic Acid-Polyglycolic Acid Copolymer , Polymers/chemistry
18.
Int J Pharm ; 439(1-2): 92-9, 2012 Dec 15.
Article in English | MEDLINE | ID: mdl-23041132

ABSTRACT

Poor water solubility of drugs can complicate their commercialisation because of reduced drug oral bioavailability. Formulation strategies such as increasing the drug surface area are frequently employed in an attempt to increase dissolution rate and hence, improve oral bioavailability. Maximising the drug surface area exposed to the dissolution medium can be achieved by loading drug onto a high surface area carrier like mesoporous silica (SBA-15). The aim of this work was to investigate the impact of altering supercritical carbon dioxide (SC-CO(2)) processing conditions, in an attempt to enhance drug loading onto SBA-15 and increase the drug's dissolution rate. Other formulation variables such as the mass ratio of drug to SBA-15 and the procedure for combining the drug and SBA-15 were also investigated. A model drug with poor water solubility, fenofibrate, was selected for this study. High drug loading efficiencies were obtained using SC-CO(2), which were influenced by the processing conditions employed. Fenofibrate release rate was enhanced greatly after loading onto mesoporous silica. The results highlighted the potential of this SC-CO(2) drug loading approach to improve the oral bioavailability of poorly water soluble drugs.


Subject(s)
Carbon Dioxide/chemistry , Silicon Dioxide/chemistry , Drug Compounding/methods , Fenofibrate/chemistry , Porosity , Solubility
19.
Cell Mol Life Sci ; 69(3): 389-404, 2012 Feb.
Article in English | MEDLINE | ID: mdl-22015612

ABSTRACT

Nanoparticles (NPs) comprised of nanoengineered complexes are providing new opportunities for enabling targeted delivery of a range of therapeutics and combinations. A range of functionalities can be included within a nanoparticle complex, including surface chemistry that allows attachment of cell-specific ligands for targeted delivery, surface coatings to increase circulation times for enhanced bioavailability, specific materials on the surface or in the nanoparticle core that enable storage of a therapeutic cargo until the target site is reached, and materials sensitive to local or remote actuation cues that allow controlled delivery of therapeutics to the target cells. However, despite the potential benefits of NPs as smart drug delivery and diagnostic systems, much research is still required to evaluate potential toxicity issues related to the chemical properties of NP materials, as well as their size and shape. The need to validate each NP for safety and efficacy with each therapeutic compound or combination of therapeutics is an enormous challenge, which forces industry to focus mainly on those nanoparticle materials where data on safety and efficacy already exists, i.e., predominantly polymer NPs. However, the enhanced functionality affordable by inclusion of metallic materials as part of nanoengineered particles provides a wealth of new opportunity for innovation and new, more effective, and safer therapeutics for applications such as cancer and cardiovascular diseases, which require selective targeting of the therapeutic to maximize effectiveness while avoiding adverse effects on non-target tissues.


Subject(s)
Drug Carriers/chemistry , Nanoparticles/chemistry , Antineoplastic Agents/administration & dosage , Antineoplastic Agents/chemistry , Cardiovascular Diseases/diagnosis , Cardiovascular Diseases/therapy , Contrast Media , Drug-Eluting Stents , Humans , Nanoparticles/therapeutic use , Neoplasms/drug therapy , Polymers/chemistry
SELECTION OF CITATIONS
SEARCH DETAIL
...