Arch. Pharm. Res. (2014) 37:69–78 DOI 10.1007/s12272-013-0284-2

REVIEW

Therapeutic applications of electrospun nanofibers for drug delivery systems Young Ju Son • Woo Jin Kim • Hyuk Sang Yoo

Received: 9 September 2013 / Accepted: 29 October 2013 / Published online: 15 November 2013 Ó The Pharmaceutical Society of Korea 2013

Abstract Electrospun nanofiber drug delivery systems have been studied using various techniques. Herein, we describe the fabrication of a drug-incorporating nanofiber. Drugs, such as proteins, peptide, antibodies, and small molecule drugs, can be loaded within or on the surface of nanofibers according to their properties. Hydrophobic drugs are directly dissolved with a polymer in an organic solvent before electrospinning. However, it is preferred to surface-immobilize bioactive molecules on nanofibers by physical absorption or chemical conjugation. Especially, chemically surface-immobilized proteins on a nanofiber mesh stimulate cell differentiation and proliferation. Using a dual electrospinning nozzle to create nanofiber sheet layers, which are stacked on top of one another, the initial burst release is reduced compared with solid nanofibers because of the layers. Furthermore, hybridization of electrospun nanofibers with nanoparticles, microspheres, and hydrogels is indirect drug loading method into the nanofibers. It is also possible to produce multi-drug delivery systems with timed programmed release. Keywords Nanofiber  Electrospinning  Drug delivery  Tissue engineering

Y. J. Son  H. S. Yoo Department of Biomedical Materials Engineering, Kangwon National University, Chuncheon 200-701, Republic of Korea W. J. Kim Department of Internal Medicine, School of Medicine, Kangwon National University, Chuncheon, Republic of Korea H. S. Yoo (&) Institute of Bioscience and Bioengineering, Kangwon National University, Chuncheon, Republic of Korea e-mail: [email protected]

Introduction Drug delivery systems using electrospun nanofibers have been developed in the last number of decades for application in many areas, such as tissue scaffolds, anti-bacterial sheets, and anti-cancer therapy (Lee et al. 2012; Tambralli et al. 2009; Wang et al. 2013). Although nanofibers alone have many advantages, such as high surface to volume ratio, porosity, and a structure that mimics the extracellular matrix (ECM) structure, the production of flexible sheets and loading of drugs has improved the function of nanofibers. Hydrophobic polymers such as poly(e-caploractone) (PCL) and poly(urethane) (PU) are easy to electrospin and exhibit high mechanical strength and elastic properties after electrospinning (Geun Hyung 2008; Lim et al. 2008). However, the hydrophobicity of electrospun nanofibers is not good for cell attachment and the nanofibers must be further modified to add hydrophilicity (Li et al. 2003). Nanofiber scaffolds have also been improved by loading proteins within/on the nanofiber mesh (Choi et al. 2008; Jia et al. 2013; Kim and Park 2006). Successful drug loading into the nanofiber for sustained release has been a key parameter for investigation. The high surface to volume ratio of the nanofiber is a unique structural feature; however, it causes an initial burst release of drugs (Verreck et al. 2003a; Kim et al. 2004; Yan et al. 2009). According to type of drug incorporated, such as small molecule drugs, proteins, and genes, the loading of the drug may require a different approach to obtain a successful release profile. Drugs can be simply mixed with the polymer solution, physically or chemically surfaceimmobilized, or indirectly loaded onto the nanofiber. The development of single nozzle to co-axial nozzle electrospinning could support the fabrication of a solid nanofiber that can control the initial burst release, overall release

123

70

Y. J. Son, H. S. Yoo

Fig. 1 Schematic diagram of simple drug loading method within the nanofiber or on the surface of the nanofiber and its release kinetics

kinetics, and multi-drug loading properties (Liao et al. 2006; Zhang et al. 2006). Furthermore, other types of biomedical devices, such as nanoparticles, microspheres, liposomes, and hydrogels can be hybridized to nanofibers to optimize the properties for specific applications (Kim and Yoo 2010; Ionescu et al. 2010; Mickova et al. 2012). This review focuses on various methods of drug loading and their advantages and limitations. We also summarize the different applications according to the methods of drug loading.

Simple drug loading In drug delivery systems, drugs can be loaded to the nanofiber by the processing of co-electrospinning or surface immobilized as shown in Fig. 1. Characteristics of drugs and the application of the device decide drug loading method. According to drug loading method within/on the nanofiber, it is expected different drug release process. Therefore, it is focused on summary two different drug incorporation skills, characteristic of drugs, and its application in this section.

123

Co-electrospinning of drugs and polymer The simplest method of drug incorporation to nanofibers is to dissolve the drugs and polymer in the same solvent and coelectrospin. The incorporation of drugs within nanofibers exhibit high drug-loading efficiency in combination with self-assembled nanoparticles or microspheres that are welldispersed within the fibers and sheets (Verreck et al. 2003b). The first report describing drug delivery from an electrospun polymer was by Kenawy et al. (2002). Poly(ethylene-covinyl acetate) (PEVA) and poly(lactic acid) (PLA) were coelectrospun with tetracycline hydrochloride to fabricate a tetracycline-PEVA monolithic fiber for controlled release of tetracycline. The correct concentration of PEVA/PLA blending trapped tetracycline within and improved the initial burst release. However, over 50 % of drugs were released out within 5 days. Numerous drugs have been co-electrospun, including small molecule drugs, antibiotics, and proteins (Zeng et al. 2005a; Pattama et al. 2006; Kim et al. 2004). Drug solubility in the polymer solution is one of the main factors deciding the drug loading method (Zeng et al. 2005b). Hydrophobic drugs, such as doxorubicin and paclitaxel have electrospun well with organic solvents (Xu et al.

Electrospun nanofibers for drug delivery systems

2008, 2009) Hydrophilic drugs, such as peptides and proteins have been dissolved in aqueous phase with poly(vinyl alchol) (PVA) or PEO and uniformly electrospun (Gatti et al. 2013; Tang et al. 2012). However, it was proposed that the exposure of drugs to organic solvents and high voltage, can be harmful for drugs especially bioactive molecules (Yang et al. 2008). Furthermore, most simple physical mixing drug-loading methods exhibit short release profiles within several days (Meng et al. 2011; Luong-Van et al. 2006; Yu et al. 2013). This is because drug release in the initial stages is a result of simple diffusion, while in the later stages it is polymer degradation. In particular, drugs located around the nanofiber surface are released in the burst phase owing to the high surface-tovolume feature of the nanofibrous structure. Surface immobilization on the nanofiber Physical and chemical surface immobilization of drugs can avoid drug denaturation caused by high voltage or organic solvents, while taking advantage of the unique feature of the high surface to volume ratio of nanofibers. It is also possible to control the amount of drugs immobilized by controlling the by drug feeding ratio. During physical absorption, electrostatic interactions, hydrophobic interactions, hydrogen bonding, and van der Waals interactions are the main forces that retain the drugs on the nanofiber (Yoshida et al. 2006). Heparin, a negatively-charged polysaccharide, has a strong binding affinity for growth factors, such as basic fibroblast growth factor (bFGF), vascular endothelial growth factor (EGF), transforming growth factor-b (TGF-b), and heparin-binding epidermal growth factor (HBEGF) (Rajangam et al. 2006; Yayon et al. 1991; Lee et al. 2007; Sasisekharan et al. 1997). Therefore, heparin is useful for physically immobilizing growth factors on the surface of nanofiber meshes. PCL/ gelatin nanofibers were surface-immobilized with heparin bound platelet-derived growth factor-BB (PDGF-BB) (Lee et al. 2012). First, heparin was chemically immobilized to the surface-exposed amine of PCL/gelatin nanofiber, following which PDGF-BB was trapped by heparin on the nanofiber. As a result, the PDGF-BB release kinetics from heparin-immobilized nanofibers was dramatically reduced compared with the non-heparin-treated nanofibers, and was continuously released for 20 days. Thus, heparin conjugation controlled the release kinetics of PDGF-BB. Chemical modification of the nanofiber surface with amine, carboxyl, hydroxyl, or thiol groups, is another method for immobilizing drugs. Hydrophilic polymers, such as poly(ethyleneglycol) (PEG) and poly(ethyleneimine) (PEI) were co-electrospun with PCL and PU to maintain the mechanical strength of the mesh and provide the mesh with functional groups. Chemical conjugation methods are better

71

for finely controlling the amount of incorporated drug on the nanofiber mesh than physically trapping the drug on the surface, and exhibits slow drug release kinetics with reduced initial burst release (Cho et al. 2010). Successful loading and release of a drug using functionalized hydroxyl polymer on nanofibers was introduced as a gene delivery system. Kim and Yoo (2010, 2013a, c) developed a gene-incorporated electrospun nanofiber mesh for wound healing. PCL-PEG nanofiber surface was modified with linear PEI to the end of PEG via a matrix metalloproteinase (MMP)-cleavable peptide, which was cleaved by MMPs in wound site. Plasmid DNA or small interfering RNA (siRNA) chemically conjugated to positively-charged amine PEI on the surface of the nanofiber according to the N/P ratio exhibited a successful release profile and gene transfection in the wound site. Surface-immobilized molecules do not always need to be released from the matrix. The ECM mimicking structure of nanofibers, whether random or aligned, have been studied for the purpose of stem cell differentiation into osteoblasts and neurons, even without any induction reagents (Ghoroghi et al. 2013; Schofer et al. 2009; Fang et al. 2010; Zhang et al. 2012a). The addition of bioactive molecules immobilized on the scaffold could enhance these effects (Pritchard et al. 2010; Jeong et al. 2010). Proteins, peptides, antibodies, and carbohydrates have been fixed on scaffold via chemical conjugation to surface exposed functional groups, such as carboxylate, thiol, and primary amine groups (Chan et al. 2009; Chua et al. 2005; Kim and Park 2006; Tigli et al. 2010). Cho et al. surface-immobilized nerve growth factor (NGF) on aligned nanofibers for the neuronal differentiation of mesenchymal stem cells (MSCs). Chemically-immobilized NGF was not released from the mesh over 7 days, and MSC differentiation into neuronal cells was effective owing to the synergic effect of the aligned nanofibrous structure and immobilized NGF on the nanofibers. Sheath nanofiber The limitation of drug release kinetic of the nanofiber with simple drug loading method is possible to be improved by introducing sheath layer on the solid fiber or mesh as shown in Fig. 2. Co-axial electrospinning technique has been developed for core/sheath nanofiber with drug within core of the nanofiber. Also, multi-layer stacked nanofiber was developed with subsequent electrospinning of two different solutions. Co-axial nanofiber Electrospinning can be modified for loading multiple drugs into a device and individually control the release kinetics of each drug. A co-axial needle is a horizontal arrangement

123

72

Y. J. Son, H. S. Yoo

Fig. 2 Schematic diagram of drug loaded nanofiber with sheath layer and its drug release kinetics

of outer and inner needles that separate two different solutions. It is easy to electrospin two immiscible polymer solutions containing drugs in the core and sheath because the two components are simultaneously electrospun from separate capillaries (Wang et al. 2012; Yu et al. 2011). Most of the core/sheath nanofibers used in drug delivery systems are loaded with proteins in core by co-electrospinning with the inner solution (Yu et al. 2013; Kim et al. 2008b; Song et al. 2013). In comparison to co-electrospinning of a drug with a single nozzle, the drug-loading efficiency is still high and the initial burst release property is decreased using sheaths of nanofibers. Hydrophobic shell materials, such as slow-degrading PCL, act as a barrier to simple diffusion of a core-loaded drug (Song et al. 2013). Drug release occurs after swelling or dissolving of the core polymer resulting in the formation of pores in the shell after dissolution of hydrophilic portion in core (Liao et al. 2006). Multi-drug delivery systems are also possible using co-axial electrospinning. Various drugs can be loaded by the co-axial system using two drug-loading methods, coelectrospinning by simple mixing and chemical surface modification, as explained in the previous section. Therefore, using two different methods the scaffold exhibits two independent drug release kinetics. In our previous study, we developed two different growth factor-loaded co-axial nanofibers (Choi and Yoo 2010b). The inner component

123

was fabricated by electrospinning in the aqueous phase with bFGF to prevent denaturation of the protein in the organic solvent of the outer solution. After electrospinning, EGF was chemically conjugated to surface exposed amines on the mesh. Drug release kinetics of the core/shell nanofiber exhibit reduced initial burst release of core-loaded drug by prevention of the initial diffusion of the drug by the shell layer, which was maximized up to 90 % in 7 days. Furthermore, shell immobilized protein was rarely released. Therefore, released bFGF and surface-immobilized EGF stimulated enhanced cell viability compared with bFGF-loaded nanofibers or EGF surface-immobilized nanofibers alone. Layer-by-layer nanofibers A layer-by-layer (LBL) system has been applied to many types of biomedical devices, nanoparticles, films, and micelles (Krishnan et al. 2007; Hettiarachchi et al. 2009; Li et al. 2005; Galyean et al. 2009; Park et al. 2012). Many studies showed reduced initial burst with zero-order release kinetics using LBL devices (Poon et al. 2011; Shutava et al. 2009; Kim et al. 2008a; De Geest et al. 2006). The system was applied to electrospun nanofibers by stacking nanofiber sheets as shown in Fig. 2 (Kidoaki et al. 2005). This can control drug release by sheet barriers, similar to co-axial

Electrospun nanofibers for drug delivery systems

73

Fig. 3 Schematic diagram of hybrid system of the nanofiber with nano-and micro-sized devices and hydrogel based drug delivery system, and its release kinetics

nanofibers. There are several studies investigating LBL nanofibers using the stacking of sheets and deposition of coating materials to monolithic fibers (Yang et al. 2009; Nogueira et al. 2010; Lee et al. 2009). Multi-layer stacked nanofibers were introduced into the fabrication of multicell layers for three-dimensional tissue scaffolds, cell filtration, and drug delivery systems (Yang et al. 2009; Pham et al. 2006; Fathi-Azarbayjani and Chan 2010). Okuda et al. (2010) applied LBL nanofibers to time programmed dual-drug delivery system. The drug loading method was the co-electrospinning of a drug and polymer mixture. Sequential electrospinning of different solutions could fabricate a multilayered electrospun nanofiber mesh. Two kinds of poly(L-lactide-co-e-caploractone) (PLCL) solutions containing two different drugs were prepared and sequentially electrospun to fabricate nanofiber meshes with a time-programmed dual release function. PLCL polymer solution without drugs was also electrospun between the two different drug-loaded solution electrospinning steps to form a barrier for the purpose of time-controlled drug release. Therefore, the first drug-loaded layer was exposed on the top of the device and the second drug-loaded nanofiber sheet was sandwiched between the bottom and barrier layers. As the result, drugs loaded in first layer were released earlier than drugs loaded in second layer fabricated by the same drug-loading method of physical mixture co-electrospinning.

Hybrid system of nanofibers The electrospun nanofiber based drug delivery system have been hybridized with other type of devices, nano- and micro-sized carrier and hydrogel for efficient drug delivery and extended application of the mesh as shown in Fig. 3. Drug loading and release kinetics of the hybrid system and its advantages to the application was introduced in this section. Nanofiber with nano- and micro-sized devices For the improvement of drug-loading efficiency, release pattern, drug safety, and better functionality of the device, electrospun nanofibers have been hybridized with other type of devices. Nano- and micro-sized biomedical devices, such as polymeric nanoparticles, nanotubes, micelles, microspheres, and liposomes have been introduced into nanofibers for a controlled drug release system (Son and Yoo 2012; Liwen et al. 2008; Ionescu et al. 2010; Shao et al. 2011). Nano and micro-particulate drug embedded nanofibers are similar to co-axial nanofibers as they protect drug stability from organic solvents in the fabrication stage and prolong the period of drug delivery (Yoo et al. 2009). Moreover, it is an easier method than co-axial electrospinning because it uses a single nozzle. The development of multi-drug loading systems with nanofibers is more

123

74

flexible with the hybridization of nanoparticles and nanofibers. Multi-drugs can be loaded into nanoparticles and nanofibers by co-electrospinning (Wang et al. 2010), and two different types of nanoparticles with different drugs can be electrospun with the polymer solution (Jo et al. 2009). Furthermore, drugs may not only be incorporated by co-electrospinning, but surface-immobilization is also possible in this system. In our group, micellized drugs were introduced on the surface of nanofibers by a surface-immobilized polymer chain (Chen et al. 2007; Son and Yoo 2012). A thermo-sensitive polymer, Pluronic F127, was used to modify the nanofiber surface of electrospun PCL-PEG and encapsulate the model drug, micellized dexamethasone. It showed that up to 98 % drugloading efficiency by the largest amount of Pluronic chain on the nanofiber and suppressed the release profile for 30 days. Even though surface immobilization is a safe method for drug stability, the amount of drugs being loaded into nanofibers is limited by the area of the nanofiber sheet. Nanofibers and hydrogels Hydrogels were introduced into nanofiber scaffolds because of their superior water retention, dual drug delivery or as a backbone device for the nanofiber in tissue engineering. The importance of extending the variety of nanofiber scaffold shapes was recognized and studies focused on micrometer scale nanofibrous hydrogels, which can be easily implanted inside the body (Lee et al. 2011, 2013a). PEG solution was cross-linked by UV on electrospun PCL nanofibers to obtain two different domains in the nanofiberbound hydrogel. For the proposed application of protein delivery, bovine serum albumin (BSA) was homogeneously loaded into nanofibers as a model protein. The BSAhybridized nanofibers localized in the hydrogel dramatically reduced the initial burst release because the hydrogel delayed the diffusion rate of BSA after released from the PCL nanofibers and the release was maintained for 30 days. Hydrogels have not only been combined with the nanofiber layer, but the hydrogel property has been directly introduced into electrospun nanofibers. Hydrogel-type materials are preferable over conventional hydrophobic materials in tissue engineering when considering cell adhesion and proliferation (Loh et al. 2010). Therefore, the hybridization of a hydrogel to a hydrophobic nanofiber improved the potency of the nanofiber in tissue engineering. The key point of hydrogel nanofibers is the electrospun polymer should maintain its fibrous structure in water with the hydrogel properties. However, hydrogel nanofibers exhibited high initial burst release of loaded drugs because of swelling. Surface treatment can reduce the initial burst of the nanofiber. PVA, which is a hydrophilic, biodegradable, and biocompatible polymer, was co-electrospun with

123

Y. J. Son, H. S. Yoo

drugs and surface-modified with fluorine to control the release period and rate (Im et al. 2010). According to the fluorine gas pressure treatment to the nanofiber surface, the drug release rate for 67 days was changed and even reduced the burst release during the initial stage because fluorination reduced the swelling property of PVA fibers in the aqueous phase. However, exposure of the nanofiber to extreme conditions, such as high temperature for fluorination, changes the morphology of nanofiber to a film, which would be not ideal for a biomolecule delivery device. Introducing hydrophilic chains into a hydrophobic polymer was performed and electrospun to produce a highly water-swellable nanofiber drug delivery system. Application Electrospun nanofiber as sustained drug delivery device has been mainly applied to tissue engineering. According to the purposes, the nanofiber can be a drug delivery carrier or a scaffold. As we explained in previous sections, incorporated drugs in/on nanofiber is not always released out. Surface immobilized proteins, peptides, other bioactive molecules are mainly related to cell adhesiveness and differentiation (Lee et al. 2013b; Choi et al. 2013). As a drug delivery device, mesh structure incorporating drugs have been targeted to antibacterial patch, mucosa, colon, wound site on skin, and cardiac tissue for local delivery system (Tian et al. 2012; Li et al. 2013; Yohe et al. 2012; Choi and Yoo 2010a). Patch type of drug loaded nanofiber is efficient device (Aduba et al. 2013). Electrospun nanofiber shows high mucoadhesive strength depending on composed polymers, and highly interacted with mucosal epithelial cells (Sharma et al. 2013). Also, sustained release of growth factors, DNAs, and siRNAs for a long period at wound site showed successful wound healing process (Kim and Yoo 2010, 2013a, b). As a scaffold of cells/tissue, ECM mimicking structure of the nanofibers is the main parameter to affect to cell differentiation. However, it was required to raise up cell attachment ratio to electrospun nanofiber composed of hydrophobic synthetic polymer such as PLGA, PCL, PLA, PU, and etc. that have advantages of proper mechanical strength and easy materials to be electrospun. Modification of the nanofiber with bioactive molecules surface can solve this problem. PLGA/PLGA-b-PEG-NH2 was prepared by electrospinning and a cell adhesive peptide, Arg–Gly–Asp (RGD) was immobilized on the surface for a potential wound dressing device (Kim and Park 2006). As the result, the surface property was changed to be hydrophilic, and NIH3T3 fibroblast successfully attached and proliferated a confluence state on RGD immobilized nanofiber for 5 days. Recently, the application of drug surface modified nanofiber was extended to stem cell differentiation;

Electrospun nanofibers for drug delivery systems

pluripotent stem cells, mesenchymal stem cell, and embryonic stem cell (Ren et al. 2013; Venugopal et al. 2012; Smith Callahan et al. 2013; Kolambkar et al. 2013). More interestingly, surface immobilized biomolecule technique has been improved to give gradients of incorporated drug on a nanofiber sheet to control stem cell differentiation (Zhang et al. 2012b; Zhu et al. 2012).

Conclusion The drug delivery system of electrospun nanofibers has been developed with synergic effects with the structural features of nanofibers. Therefore, various type of electrospun nanofiber were examined and grouped according to their drug-loading method. Most other literatures mainly distinguish electrospun nanofibers by co-electrospinning and chemical surface immobilization, which details direct and indirect drug incorporation into nanofibers and their hybridization system. According to the drug properties, such as hydrophobicity and hydrophilicity, bioactive molecules, and easy denaturation, we then differentiated the drug-loading abilities. With the development of many types of drug-loading methods, the initial burst release can be reduced, and zero-order release has also been exhibited (Immich et al. 2013; Zhang et al. 2006). Furthermore, hybridization with other types of biomedical devices elevated not only the drug release kinetics, but also produced scaffolds that can be applied as implantable tissue scaffold using hydrogel nanofibers. With many of advantages of nanofibers, such as flexible material selectivity, drugloading methods, variety of structural architectures and ECM mimicking structure, a massive variety of applications of nanofibers have been achieved, especially in tissue engineering. Released drugs from the nanofibers have been shown to control cellular metabolism, such as proliferation and differentiation (Choi and Yoo 2010a; Chen et al. 2012). They have also been shown to increase efficiency in local gene delivery systems. Furthermore, some meshes that have been modified with surface-immobilized proteins that are not released can be applied as cell sheets for attachment and differentiation. Therefore, it is expected that further applications of nanofibers with different drug combinations will be investigated and the advantages of the nanofibrous structural features will be exploited. Acknowledgments This work was supported by a grant from the National R&D Program for Cancer Control, Ministry for Health and Welfare, Republic of Korea.

References Aduba Jr, D.C., J.A. Hammer, Q. Yuan, W. Andrew Yeudall, G.L. Bowlin, and H. Yang. 2013. Semi-interpenetrating network

75 (sIPN) gelatin nanofiber scaffolds for oral mucosal drug delivery. Acta Biomaterialia 9: 6576–6584. Chan, C.K., S. Liao, B. Li, R.R. Lareu, J.W. Larrick, S. Ramakrishna, and M. Raghunath. 2009. Early adhesive behavior of bonemarrow-derived mesenchymal stem cells on collagen electrospun fibers. Biomedical Materials 4: 035006–035016. Chen, C., G. Lv, C. Pan, M. Song, C. Wu, D. Guo, X. Wang, B. Chen, and Z. Gu. 2007. Poly(lactic acid) (PLA) based nanocomposites—a novel way of drug-releasing. Biomedical Materials 2: L1–L4. Chen, M., S. Gao, M. Dong, J. Song, C. Yang, K.A. Howard, J. Kjems, and F. Besenbacher. 2012. Chitosan/siRNA nanoparticles encapsulated in PLGA nanofibers for siRNA delivery. ACS Nano 6: 4835–4844. Cho, Y.I., J.S. Choi, S.Y. Jeong, and H.S. Yoo. 2010. Nerve growth factor (NGF)-conjugated electrospun nanostructures with topographical cues for neuronal differentiation of mesenchymal stem cells. Acta Biomaterialia 6: 4725–4733. Choi, J.S., K.W. Leong, and H.S. Yoo. 2008. In vivo wound healing of diabetic ulcers using electrospun nanofibers immobilized with human epidermal growth factor (EGF). Biomaterials 29: 587–596. Choi, J.S., P.B. Messersmith, and H.S. Yoo. 2013. Decoration of electrospun nanofibers with monomeric catechols to facilitate cell adhesion. Macromolecular Bioscience. doi:10.1002/mabi. 201300281. Choi, J.S., and H.S. Yoo. 2010a. Nano-inspired fibrous matrix with bi-phasic release of proteins. Journal of Nanoscience and Nanotechnology 10: 3038–3045. Choi, J.S., and H.S. Yoo. 2010b. Pluronic/chitosan hydrogels containing epidermal growth factor with wound-adhesive and photo-crosslinkable properties. Journal of Biomedical Materials Research Part A 95: 564–573. Chua, K.N., W.S. Lim, P. Zhang, H. Lu, J. Wen, S. Ramakrishna, K.W. Leong, and H.Q. Mao. 2005. Stable immobilization of rat hepatocyte spheroids on galactosylated nanofiber scaffold. Biomaterials 26: 2537–2547. De Geest, B.G., C. De´jugnat, E. Verhoeven, G.B. Sukhorukov, A.M. Jonas, J. Plain, J. Demeester, and S.C. De Smedt. 2006. Layerby-layer coating of degradable microgels for pulsed drug delivery. Journal of Controlled Release 116: 159–169. Fang, R., E. Zhang, L. Xu, and S. Wei. 2010. Electrospun PCL/PLA/ HA based nanofibers as scaffold for osteoblast-like cells. Journal of Nanoscience and Nanotechnology 10: 7747–7751. Fathi-Azarbayjani, A., and S.Y. Chan. 2010. Single and multi-layered nanofibers for rapid and controlled drug delivery. Chemical and Pharmaceutical Bulletin (Tokyo) 58: 143–146. Galyean, A.A., R.W. Day, J. Malinowski, K.W. Kittredge, and M.C. Leopold. 2009. Polyelectrolyte-linked film assemblies of nanoparticles and nanoshells: Growth, stability, and optical properties. Journal of Colloid and Interface Science 331: 532–542. Gatti, J.W., M.C. Smithgall, S.M. Paranjape, R.J. Rolfes, and M. Paranjape. 2013. Using electrospun poly(ethylene-oxide) nanofibers for improved retention and efficacy of bacteriolytic antibiotics. Biomedical Microdevices 15: 887–893. Geun Hyung, K. 2008. Electrospun PCL nanofibers with anisotropic mechanical properties as a biomedical scaffold. Biomedical Materials 3: 025010. Ghoroghi, F.M., L.B. Hejazian, B. Esmaielzade, M. Dodel, M. Roudbari, and M. Nobakht. 2013. Evaluation of the effect of NT-3 and biodegradable poly-L-lactic acid nanofiber scaffolds on differentiation of rat hair follicle stem cells into neural cells in vitro. Journal of Molecular Neuroscience. doi:10.1007/ s12031-12013-10073-x. Hettiarachchi, K., S. Zhang, S. Feingold, A.P. Lee, and P.A. Dayton. 2009. Controllable microfluidic synthesis of multiphase drug-

123

76 carrying lipospheres for site-targeted therapy. Biotechnology Progress 25: 938–945. Im, J.S., J. Yun, Y.M. Lim, H.I. Kim, and Y.S. Lee. 2010. Fluorination of electrospun hydrogel fibers for a controlled release drug delivery system. Acta Biomaterialia 6: 102–109. Immich, A.P., M.L. Arias, N. Carreras, R.L. Boemo, and J.A. Tornero. 2013. Drug delivery systems using sandwich configurations of electrospun poly(lactic acid) nanofiber membranes and ibuprofen. Materials Science and Engineering C: Materials for Biological Applications 33: 4002–4008. Ionescu, L.C., G.C. Lee, B.J. Sennett, J.A. Burdick, and R.L. Mauck. 2010. An anisotropic nanofiber/microsphere composite with controlled release of biomolecules for fibrous tissue engineering. Biomaterials 31: 4113–4120. Jeong, S.I., M.D. Krebs, C.A. Bonino, S.A. Khan, and E. Alsberg. 2010. Electrospun alginate nanofibers with controlled cell adhesion for tissue engineering. Macromolecular Bioscience 10: 934–943. Jia, L., M. Prabhakaran, X. Qin, D. Kai, and S. Ramakrishna. 2013. Biocompatibility evaluation of protein-incorporated electrospun polyurethane-based scaffolds with smooth muscle cells for vascular tissue engineering. Journal of Materials Science 48: 5113–5124. Jo, E., S. Lee, K.T. Kim, Y.S. Won, H.-S. Kim, E.C. Cho, and U. Jeong. 2009. Core-sheath nanofibers containing colloidal arrays in the core for programmable multi-agent delivery. Advanced Materials 21: 968–972. Kenawy, E.-R., G.L. Bowlin, K. Mansfield, J. Layman, D.G. Simpson, E.H. Sanders, and G.E. Wnek. 2002. Release of tetracycline hydrochloride from electrospun poly(ethylene-covinylacetate), poly(lactic acid), and a blend. Journal of Controlled Release 81: 57–64. Kidoaki, S., I.K. Kwon, and T. Matsuda. 2005. Mesoscopic spatial designs of nano- and microfiber meshes for tissue-engineering matrix and scaffold based on newly devised multilayering and mixing electrospinning techniques. Biomaterials 26: 37–46. Kim, B.S., S.W. Park, and P.T. Hammond. 2008a. Hydrogen-bonding layer-by-layer-assembled biodegradable polymeric micelles as drug delivery vehicles from surfaces. ACS Nano 2: 386–392. Kim, G.H., T. Min, S.A. Park, and W.D. Kim. 2008b. Coaxially electrospun micro/nanofibrous poly(epsilon-caprolactone)/eggshell-protein scaffold. Bioinspiration and Biomimetics 3: 016006. Kim, H.S., and H.S. Yoo. 2010. MMPs-responsive release of DNA from electrospun nanofibrous matrix for local gene therapy: in vitro and in vivo evaluation. Journal of Controlled Release 145: 264–271. Kim, H.S., and H.S. Yoo. 2013a. In vitro and in vivo epidermal growth factor gene therapy for diabetic ulcers with electrospun fibrous meshes. Acta Biomaterialia 9: 7371–7380. Kim, H.S., and H.S. Yoo. 2013b. Matrix metalloproteinase-inspired suicidal treatments of diabetic ulcers with siRNA-decorated nanofibrous meshes. Gene Therapy 20: 378–385. Kim, H.S., and H.S. Yoo. 2013c. Matrix metalloproteinase-inspired suicidal treatments of diabetic ulcers with siRNA-decorated nanofibrous meshes. Gene Therapy 20: 378–385. Kim, K., Y.K. Luu, C. Chang, D.F. Fang, B.S. Hsiao, B. Chu, and M. Hadjiargyrou. 2004. Incorporation and controlled release of a hydrophilic antibiotic using poly(lactide-co-glycolide)-based electrospun nanofibrous scaffolds. Journal of Controlled Release 98: 47–56. Kim, T.G., and T.G. Park. 2006. Biomimicking extracellular matrix: cell adhesive RGD peptide modified electrospun poly(D,L-lacticco-glycolic acid) nanofiber mesh. Tissue Engineering 12: 221–233. Kolambkar, Y.M., M. Bajin, A. Wojtowicz, D.W. Hutmacher, A.J. Garcia, and R.E. Guldberg. 2013. Nanofiber orientation and

123

Y. J. Son, H. S. Yoo surface functionalization modulate human mesenchymal stem cell behavior in vitro. Tissue Engineering, Parts A. doi:10.1089/ ten.TEA.2012.0426. Krishnan, R.S., M.E. Mackay, P.M. Duxbury, A. Pastor, C.J. Hawker, B. Van Horn, S. Asokan, and M.S. Wong. 2007. Self-assembled multilayers of nanocomponents. Nano Letters 7: 484–489. Lee, H.J., H.S. Kim, H.O. Kim, and W.G. Koh. 2011. Micropatterns of double-layered nanofiber scaffolds with dual functions of cell patterning and metabolite detection. Lab on a Chip 11: 2849–2857. Lee, H.J., Y.H. Park, and W.-G. Koh. 2013a. Fabrication of nanofiber microarchitectures localized within hydrogel microparticles and their application to protein delivery and cell encapsulation. Advanced Functional Materials 23: 591–597. Lee, J., J.J. Yoo, A. Atala, and S.J. Lee. 2012. The effect of controlled release of PDGF-BB from heparin-conjugated electrospun PCL/ gelatin scaffolds on cellular bioactivity and infiltration. Biomaterials 33: 6709–6720. Lee, J.A., K.C. Krogman, M. Ma, R.M. Hill, P.T. Hammond, and G.C. Rutledge. 2009. Highly reactive multilayer-assembled TiO2 coating on electrospun polymer nanofibers. Advanced Materials 21: 1252–1256. Lee, J.S., D.H. Go, J.W. Bae, S.J. Lee, and K.D. Park. 2007. Heparin conjugated polymeric micelle for long-term delivery of basic fibroblast growth factor. Journal of Controlled Release 117: 204–209. Lee, Y.J., J.-H. Lee, H.-J. Cho, H.K. Kim, T.R. Yoon, and H. Shin. 2013b. Electrospun fibers immobilized with bone forming peptide-1 derived from BMP7 for guided bone regeneration. Biomaterials 34: 5059–5069. Li, M., E. Dujardin, and S. Mann. 2005. Programmed assembly of multi-layered protein/nanoparticle-carbon nanotube conjugates. Chemical Communications 21: 4952–4954. Li, W.-J., K.G. Danielson, P.G. Alexander, and R.S. Tuan. 2003. Biological response of chondrocytes cultured in three-dimensional nanofibrous poly(e-caprolactone) scaffolds. Journal of Biomedical Materials Research Part A 67A: 1105–1114. Li, X., M.A. Kanjwal, L. Lin, and I.S. Chronakis. 2013. Electrospun polyvinyl-alcohol nanofibers as oral fast-dissolving delivery system of caffeine and riboflavin. Colloids and Surfaces B: Biointerfaces 103: 182–188. Liao, I.C., S.Y. Chew, and K.W. Leong. 2006. Aligned core-shell nanofibers delivering bioactive proteins. Nanomedicine 1: 465–471. Lim, C.T., E.P.S. Tan, and S.Y. Ng. 2008. Effects of crystalline morphology on the tensile properties of electrospun polymer nanofibers. Applied Physics Letters 92: 1419080–1419083. Liwen, J., S. Carl, A.K. Saad, and Z. Xiangwu. 2008. Preparation and characterization of silica nanoparticulate–polyacrylonitrile composite and porous nanofibers. Nanotechnology 19: 085605. Loh, X.J., P. Peh, S. Liao, C. Sng, and J. Li. 2010. Controlled drug release from biodegradable thermoresponsive physical hydrogel nanofibers. Journal of Controlled Release 143: 175–182. Luong-Van, E., L. Grøndahl, K.N. Chua, K.W. Leong, V. Nurcombe, and S.M. Cool. 2006. Controlled release of heparin from poly(ecaprolactone) electrospun fibers. Biomaterials 27: 2042–2050. Meng, Z.X., W. Zheng, L. Li, and Y.F. Zheng. 2011. Fabrication, characterization and in vitro drug release behavior of electrospun PLGA/chitosan nanofibrous scaffold. Materials Chemistry and Physics 125: 606–611. Mickova, A., M. Buzgo, O. Benada, M. Rampichova, Z. Fisar, E. Filova, M. Tesarova, D. Lukas, and E. Amler. 2012. Core/shell nanofibers with embedded liposomes as a drug delivery system. Biomacromolecules 13: 952–962. Nogueira, G.M., A.J. Swiston, M.M. Beppu, and M.F. Rubner. 2010. Layer-by-layer deposited chitosan/silk fibroin thin films with anisotropic nanofiber alignment. Langmuir 26: 8953–8958.

Electrospun nanofibers for drug delivery systems Okuda, T., K. Tominaga, and S. Kidoaki. 2010. Time-programmed dual release formulation by multilayered drug-loaded nanofiber meshes. Journal of Controlled Release 143: 258–264. Park, J.H., H.A. Kim, S.H. Cho, and M. Lee. 2012. Characterization of hydrophobic anti-cancer drug-loaded amphiphilic peptides as a gene carrier. Journal of Cellular Biochemistry 113: 1645–1653. Pattama, T., R. Uracha, and S. Pitt. 2006. Drug-loaded electrospun mats of poly(vinyl alcohol) fibres and their release characteristics of four model drugs. Nanotechnology 17: 2317. Pham, Q.P., U. Sharma, and A.G. Mikos. 2006. Electrospun poly(epsilon-caprolactone) microfiber and multilayer nanofiber/ microfiber scaffolds: characterization of scaffolds and measurement of cellular infiltration. Biomacromolecules 7: 2796–2805. Poon, Z., D. Chang, X. Zhao, and P.T. Hammond. 2011. Layer-bylayer nanoparticles with a pH-sheddable layer for in vivo targeting of tumor hypoxia. ACS Nano 5: 4284–4292. Pritchard, C.D., K.M. Arner, R.A. Neal, W.L. Neeley, P. Bojo, E. Bachelder, J. Holz, N. Watson, E.A. Botchwey, R.S. Langer, and F.K. Ghosh. 2010. The use of surface modified poly(glycerol-cosebacic acid) in retinal transplantation. Biomaterials 31: 2153–2162. Rajangam, K., H.A. Behanna, M.J. Hui, X. Han, J.F. Hulvat, J.W. Lomasney, and S.I. Stupp. 2006. Heparin binding nanostructures to promote growth of blood vessels. Nano Letters 6: 2086–2090. Ren, Y.-J., S. Zhang, R. Mi, Q. Liu, X. Zeng, M. Rao, A. Hoke, and H.-Q. Mao. 2013. Enhanced differentiation of human neural crest stem cells towards the Schwann cell lineage by aligned electrospun fiber matrix. Acta Biomaterialia 9: 7727–7736. Sasisekharan, R., S. Ernst, and G. Venkataraman. 1997. On the regulation of fibroblast growth factor activity by heparin-like glycosaminoglycans. Angiogenesis 1: 45–54. Schofer, M.D., U. Boudriot, I. Leifeld, R.I. Sutterlin, M. Rudisile, J.H. Wendorff, A. Greiner, J.R. Paletta, and S. Fuchs-Winkelmann. 2009. Characterization of a PLLA-collagen I blend nanofiber scaffold with respect to growth and osteogenic differentiation of human mesenchymal stem cells. Scientific World Journal 9: 118–129. Shao, S., L. Li, G. Yang, J. Li, C. Luo, T. Gong, and S. Zhou. 2011. Controlled green tea polyphenols release from electrospun PCL/ MWCNTs composite nanofibers. International Journal of Pharmaceutics 421: 310–320. Sharma, A., A. Gupta, G. Rath, A. Goyal, R.B. Mathur, and S.R. Dhakate. 2013. Electrospun composite nanofiber-based transmucosal patch for anti-diabetic drug delivery. Journal of Materials Chemistry B 1: 3410–3418. Shutava, T.G., S.S. Balkundi, P. Vangala, J.J. Steffan, R.L. Bigelow, J.A. Cardelli, D.P. O’neal, and Y.M. Lvov. 2009. Layer-bylayer-coated gelatin nanoparticles as a vehicle for delivery of natural polyphenols. ACS Nano 3: 1877–1885. SmithCallahan, L.A., S. Xie, I.A. Barker, J. Zheng, D.H. Reneker, A.P. Dove, and M.L. Becker. 2013. Directed differentiation and neurite extension of mouse embryonic stem cell on aligned poly(lactide) nanofibers functionalized with YIGSR peptide. Biomaterials 34: 9089–9095. Son, Y.J., and H.S. Yoo. 2012. Dexamethasone-incorporated nanofibrous meshes for antiproliferation of smooth muscle cells: Thermally induced drug-loading strategy. Journal of Biomedical Materials Research Part A 100A: 2678–2685. Song, W., X. Yu, D.C. Markel, T. Shi, and W. Ren. 2013. Coaxial PCL/PVA electrospun nanofibers: osseointegration enhancer and controlled drug release device. Biofabrication 5: 035006. Tambralli, A., B. Blakeney, J. Anderson, M. Kushwaha, A. Andukuri, D. Dean, and H.W. Jun. 2009. A hybrid biomimetic scaffold composed of electrospun polycaprolactone nanofibers and self-

77 assembled peptide amphiphile nanofibers. Biofabrication 1: 025001. Tang, C., A.E. Ozcam, B. Stout, and S.A. Khan. 2012. Effect of pH on protein distribution in electrospun PVA/BSA composite nanofibers. Biomacromolecules 13: 1269–1278. Tian, L., M. Prabhakaran, X. Ding, D. Kai, and S. Ramakrishna. 2012. Emulsion electrospun vascular endothelial growth factor encapsulated poly(l-lactic acid-co-e-caprolactone) nanofibers for sustained release in cardiac tissue engineering. Journal of Materials Science 47: 3272–3281. Tigli, R.S., N.M. Kazaroglu, I.S.B. Mav, and I.O.M. Gumusderel. 2010. Cellular behavior on epidermal growth factor (EGF)immobilized PCL/gelatin nanofibrous scaffolds. Journal of Biomaterials Science, Polymer Edition 22(1–3): 207–223. Venugopal, J., R. Rajeswari, M. Shayanti, S. Low, A.R. Bongso, V. Giri Dev, G. Deepika, A.T. Choon, and S. Ramakrishna. 2012. Electrosprayed hydroxyapatite on polymer nanofibers to differentiate mesenchymal stem cells to osteogenesis. Journal of Biomaterials Science, Polymer Edition 24: 170–184. Verreck, G., I. Chun, J. Peeters, J. Rosenblatt, and M.E. Brewster. 2003a. Preparation and characterization of nanofibers containing amorphous drug dispersions generated by electrostatic spinning. Pharmaceutical Research 20: 810–817. Verreck, G., I. Chun, J. Rosenblatt, J. Peeters, A.V. Dijck, J. Mensch, M. Noppe, and M.E. Brewster. 2003b. Incorporation of drugs in an amorphous state into electrospun nanofibers composed of a water-insoluble, nonbiodegradable polymer. Journal of Controlled Release 92: 349–360. Wang, S.G., X. Jiang, P.C. Chen, A.G. Yu, and X.J. Huang. 2012. Preparation of coaxial-electrospun poly[bis(p-methylphenoxy)]phosphazene nanofiber membrane for enzyme immobilization. International Journal of Molecular Sciences 13: 14136– 14148. Wang, Y., B. Wang, W. Qiao, and T. Yin. 2010. A novel controlled release drug delivery system for multiple drugs based on electrospun nanofibers containing nanoparticles. Journal of Pharmaceutical Sciences 99: 4805–4811. Wang, Y., Y. Zhang, B. Wang, Y. Cao, Q. Yu, and T. Yin. 2013. Fabrication of core–shell micro/nanoparticles for programmable dual drug release by emulsion electrospraying. Journal of Nanoparticle Research 15: 1726–1738. Xu, X., X. Chen, P.A. Ma, X. Wang, and X. Jing. 2008. The release behavior of doxorubicin hydrochloride from medicated fibers prepared by emulsion-electrospinning. European Journal of Pharmaceutics and Biopharmaceutics 70: 165–170. Xu, X., X. Chen, Z. Wang, and X. Jing. 2009. Ultrafine PEG–PLA fibers loaded with both paclitaxel and doxorubicin hydrochloride and their in vitro cytotoxicity. European Journal of Pharmaceutics and Biopharmaceutics 72: 18–25. Yan, S., L. Xiaoqiang, T. Lianjiang, H. Chen, and M. Xiumei. 2009. Poly(l-lactide-co-e-caprolactone) electrospun nanofibers for encapsulating and sustained releasing proteins. Polymer 50: 4212–4219. Yang, X., J.D. Shah, and H. Wang. 2009. Nanofiber enabled layer-bylayer approach toward three-dimensional tissue formation. Tissue Engineering Part A 15: 945–956. Yang, Y., X. Li, M. Qi, S. Zhou, and J. Weng. 2008. Release pattern and structural integrity of lysozyme encapsulated in core–sheath structured poly(DL-lactide) ultrafine fibers prepared by emulsion electrospinning. European Journal of Pharmaceutics and Biopharmaceutics 69: 106–116. Yayon, A., M. Klagsbrun, J.D. Esko, P. Leder, and D.M. Ornitz. 1991. Cell surface, heparin-like molecules are required for binding of basic fibroblast growth factor to its high affinity receptor. Cell 64: 841–848.

123

78 Yohe, S.T., V.L.M. Herrera, Y.L. Colson, and M.W. Grinstaff. 2012. 3D Superhydrophobic electrospun meshes as reinforcement materials for sustained local drug delivery against colorectal cancer cells. Journal of Controlled Release 162: 92–101. Yoo, H.S., T.G. Kim, and T.G. Park. 2009. Surface-functionalized electrospun nanofibers for tissue engineering and drug delivery. Advanced Drug Delivery Reviews 61: 1033–1042. Yoshida, M., R. Langer, A. Lendlein, and J. Lahann. 2006. From advanced biomedical coatings to multi-functionalized biomaterials. Journal of Macromolecular Science: Part C: Polymer Reviews 46: 347–375. Yu, D.G., C. Branford-White, S.W. Bligh, K. White, N.P. Chatterton, and L.M. Zhu. 2011. Improving polymer nanofiber quality using a modified co-axial electrospinning process. Macromolecular Rapid Communications 32: 744–750. Yu, D.G., X. Wang, X.Y. Li, W. Chian, Y. Li, and Y.Z. Liao. 2013. Electrospun biphasic drug release polyvinylpyrrolidone/ethyl cellulose core/sheath nanofibers. Acta Biomaterialia 9: 5665–5672. Zeng, J., A. Aigner, F. Czubayko, T. Kissel, J.H. Wendorff, and A. Greiner. 2005a. Poly(vinyl alcohol) nanofibers by electrospinning as a protein delivery system and the retardation of enzyme release by additional polymer coatings. Biomacromolecules 6: 1484–1488.

123

Y. J. Son, H. S. Yoo Zeng, J., L. Yang, Q. Liang, X. Zhang, H. Guan, X. Xu, X. Chen, and X. Jing. 2005b. Influence of the drug compatibility with polymer solution on the release kinetics of electrospun fiber formulation. Journal of Controlled Release 105: 43–51. Zhang, D., A. Tong, L. Zhou, F. Fang, and G. Guo. 2012a. Osteogenic differentiation of human placenta-derived mesenchymal stem cells (PMSCs) on electrospun nanofiber meshes. Cytotechnology 64: 701–710. Zhang, X., X. Gao, L. Jiang, and J. Qin. 2012b. Flexible generation of gradient electrospinning nanofibers using a microfluidic assisted approach. Langmuir 28: 10026–10032. Zhang, Y.Z., X. Wang, Y. Feng, J. Li, C.T. Lim, and S. Ramakrishna. 2006. Coaxial electrospinning of (fluorescein isothiocyanateconjugated bovine serum albumin)-encapsulated poly(e-caprolactone) nanofibers for sustained release. Biomacromolecules 7: 1049–1057. Zhu, Y., Z. Mao, and C. Gao. 2012. Control over the gradient differentiation of rat BMSCs on a PCL membrane with surfaceimmobilized alendronate gradient. Biomacromolecules 14: 342–349.

Therapeutic applications of electrospun nanofibers for drug delivery systems.

Electrospun nanofiber drug delivery systems have been studied using various techniques. Herein, we describe the fabrication of a drug-incorporating na...
416KB Sizes 0 Downloads 0 Views