Submitted Successfully!
To reward your contribution, here is a gift for you: A free trial for our video production service.
Thank you for your contribution! You can also upload a video entry or images related to this topic.
Version Summary Created by Modification Content Size Created at Operation
1 + 3974 word(s) 3974 2021-04-19 10:17:23 |
2 format correct Meta information modification 3974 2021-04-20 05:42:53 |

Video Upload Options

Do you have a full video?

Confirm

Are you sure to Delete?
Cite
If you have any further questions, please contact Encyclopedia Editorial Office.
Duong, V. SLNs/NLCs Preparation Method. Encyclopedia. Available online: https://encyclopedia.pub/entry/8771 (accessed on 01 July 2024).
Duong V. SLNs/NLCs Preparation Method. Encyclopedia. Available at: https://encyclopedia.pub/entry/8771. Accessed July 01, 2024.
Duong, Van-An. "SLNs/NLCs Preparation Method" Encyclopedia, https://encyclopedia.pub/entry/8771 (accessed July 01, 2024).
Duong, V. (2021, April 19). SLNs/NLCs Preparation Method. In Encyclopedia. https://encyclopedia.pub/entry/8771
Duong, Van-An. "SLNs/NLCs Preparation Method." Encyclopedia. Web. 19 April, 2021.
SLNs/NLCs Preparation Method
Edit

Solid lipid nanoparticles (SLNs) and nanostructured lipid carriers (NLCs) are alternatives to other colloidal drug delivery systems, such as liposomes, emulsions, and polymeric nanoparticles. They have been produced using numerous methods.

Solid Lipid Nanoparticles Nanostructured Lipid Carriers Drug Delivery Preparation

1. Introduction

Solid lipid nanoparticles (SLNs) and nanostructured lipid carriers (NLCs) are colloidal particles with sizes ranging from 10 to 1000 nm [1]. They offer alternatives to other colloidal drug delivery systems, such as liposomes, emulsions, and polymeric nanoparticles. SLNs and NLCs can be prepared reproducibly in the absence of toxic organic solvents using techniques such as high-pressure homogenization or high-speed stirring [2]. The solid matrices of SLNs and NLCs contribute to increase the stabilities of active ingredients [3]. Both SLNs and NLCs are considered nanosafe carriers since they are produced from physiological and biodegradable lipids (e.g., triglycerides, partial glycerides, waxes, steroids, and fatty acids) and other materials generally recognized as safe (GRAS) [2][4]. SLNs and NLCs allow entrapment of hydrophilic and hydrophobic drugs with higher entrapment efficiencies (EEs) than liposomes [5]. Furthermore, they enable controlled drug release like polymeric nanoparticles and can be coated with appropriate ligands to target specific tissues [1]. Thus, SLNs and NLCs have been used for topical, oral, intranasal, pulmonary, and parenteral applications [6].

SLNs and NLCs have been produced using numerous methods [7][8][9]. Some methods, such as high-pressure homogenization, emulsion/solvent evaporation, phase inversion, and microemulsion, have been extensively investigated and reviewed [2][10][11]. In this entry, we describe the various methods used to prepare SLNs and NLCs and detail their advantages, disadvantages, and applications for drug delivery.

2. Preparation methods

2.1. High-Pressure Homogenization Method

High-pressure homogenization (HPH) has been widely used to prepare nanoemulsions, SLNs, and NLCs. The technique is based on the reduction of droplet and particle size under extreme pressure conditions [12]. It provides an effective, reliable means of preparing SLNs and NLCs on a large-scale. For example, this method was used to prepare stavudine-loaded SLNs at batch sizes of up to 60 kg [13] and coenzyme Q10 loaded-NLCs at a throughput of 25 kg/h [14]. Initially, coarse lipid particles are prepared and then forced through a homogenizer with a narrow gap of few microns at high pressure [8]. The pressures (usually 100–2000 bar) accelerate the liquid mixtures to a high velocity of >1000 km/h, which generates high shear stresses and cavitational forces that reduce droplet sizes to nano dimensions [1]. This homogenization process is cycled until the desired droplet size and uniformity is obtained. The advantages of HPH are short production times, ease of production, organic solvent-free operation, and scale-up feasibility [14].

Two approaches are used to produce SLNs and NLCs using HPH, which are hot and cold homogenization. For hot HPH, drugs and molten lipids are mixed at temperatures typically 5–10 °C higher than the solid lipid melting point; drugs are either dissolved or homogeneously dispersed in the molten lipids [15]. Generally, the concentration of lipids in resultant SLNs and NLCs dispersion is 5–10% (w/v). Separately, an aqueous phase containing surfactants is preheated to the same temperature as the lipid melt and then added under constant stirring to produce a hot pre-emulsion. The resultant pre-emulsion is homogenized using a piston-gap homogenizer at the same temperature [16]. Generally, desired SLNs and NLCs can be obtained after 3–5 homogenization cycles at 500–1500 bars. However, increasing cycle numbers and homogenization pressure may increase particle size (PS) due to particle coalescence under highly kinetic conditions [17]. After homogenization, the nanoemulsions are cooled, which results in lipid crystallization and the formations of SLNs and NLCs. On the other hand, hot HPH has its limitations, which include temperature-induced drug degradation and drug loss to the aqueous phase during homogenization [18]. Therefore, the technique is unsuitable for heat-sensitive or hydrophilic drugs. The temperature sensitivity issue can be overcome by cold HPH (as discussed below), whereas drug loss is usually minimized using covalently bonded lipid-drug conjugates [19]

Cold HPH was developed to overcome some of the limitations of hot HPH. For cold HPH, after dissolving/dispersing drugs in molten lipids, mixtures are rapidly cooled using liquid nitrogen or dry ice. This high cooling rate provides homogeneous dispersions of drugs in lipid matrices [1]. The lipid-drug mixtures are then pulverized using a ball mill or a mortar to a PS of 50–100 µm. The lipid microparticles are suspended in cold aqueous solutions containing surfactants and then homogenized at the cold condition (e.g., 0–4 °C) usually over 5–10 cycles at 500 bars [20]. Microparticle suspensions can also be prepared in ways that avoid the laborious pulverization step. For example, a solvent diffusion method was used to prepare a coarse suspension, which was subsequently subjected to cold HPH at 0–4 °C [21][22], which minimized drug degradation associated with hot HPH [18]. In addition, cold HPH is suitable for water-soluble drugs as it suppresses drug migration from the lipid phase to the aqueous phase [1]. In addition, several modifications can be used to improve EE and DL values further. For example, pH values of the aqueous phase can be adjusted to minimize losses of drugs with pH-dependent solubility to the aqueous phase. Drug-lipid conjugates can also be used. To prepare decitabine-loaded SLNs, decitabine-stearic acid conjugates were ground into a fine powder, dispersed in a cold aqueous solution containing surfactants, and subjected to 10–14 cycles of cold HPH at a pressure of 1000 bar, at which an EE of up to 68.9% was obtained [23]. On the downside, it should be noted that cold HPH produces larger particles with a broader particle size distribution than hot HPH [1].

2.2. High-Speed Stirring and Ultra-Sonication Methods

High-speed stirring (high-shear homogenization) and ultra-sonication are widely-used dispersing techniques. High-speed stirring is one of the simplest and most cost-effective ways of producing SLNs and NLCs [18]. According to this method, lipids are first melted at high temperatures (5–10 °C higher than the melting point of solid lipids), and drugs are dissolved or dispersed homogeneously in the molten lipids. An aqueous phase containing surfactants (at the same temperature) is then added to the drug-lipid melt, and the mixture is homogeneously dispersed using a high-shear mixer. A hot oil/water (o/w) emulsion is formed due to the shear of intense turbulent eddies. SLNs and NLCs are formed by cooling these dispersions [24]. This high-speed stirring is usually followed by ultra-sonication, which breaks droplets based on the formation, growth, and implosive collapse of bubbles [25]. When ultra-sonication is performed without the high-shear mixing stage, SLNs and NLCs produced have a broad distribution [26], presumably because sonication energy is not transferred equally in the batch. High-speed stirring and ultra-sonication have been widely used in combination to achieve SLNs and NLCs dispersions with narrow particle distributions [27]. Both high-speed stirring and ultra-sonication are easy to handle and can be used widely without organic solvents [28]. However, both suffer from the disadvantage that drugs are exposed to high temperatures for extended times [29]. Moreover, the size distributions of SLNs and NLCs produced by high-speed stirring are broad and particles are micro-sized. The disadvantage of ultra-sonication is that products are contaminated with metals originating from sonicator probes [1]. Bath sonication provides a means of circumventing this problem but must be combined with other follow-on procedures to reduce PS and PDI values. Furthermore, using high-speed stirring and/or ultra-sonication methods require high surfactant, whereas total lipid concentrations are low [30].

2.3. Microemulsion Method

This method was first developed in the early 1990s and has since been investigated by several researchers [31]. The method involves diluting a microemulsion in a cold aqueous solution, which results in the formation of nanoemulsion and the subsequent formation of SLNs and NLCs by lipid precipitation. Briefly, a drug is dissolved in molten lipids at a temperature above the lipids melting point, and then an aqueous phase containing water and surfactant (pre-heated to the same temperature) is added under mild stirring to form a transparent and thermodynamically stable microemulsion [32]. The microemulsion is then poured into a cold aqueous solution (2–10 °C) under gentle mechanical mixing [18]. Typically, the volume of the cold aqueous phase is 25 to 50 times greater than that of the hot emulsion [1]. Upon dilution, a nanoemulsion is formed and lipids immediately crystallize to form SLNs or NLCs [33]. The microemulsion method is simple and reproducible and can be scaled up. For large-scale production, the microemulsion can be prepared in a large temperature-controlled tank and pumped into another tank containing cold water for lipid precipitation [18]. In addition, this method is solvent-free. Its limitations are a large amount of water required to dilute microemulsions and high surfactant usage [34]. Excess water can be removed by lyophilization or ultra-filtration to concentrate SLNs and NLCs dispersions [18].

2.4. Solvent Emulsification-Diffusion Method

The solvent emulsification-diffusion method is mainly used to produce polymeric nano-carriers. In 2003, Trotta et al. first used this technique to prepare SLNs and NLCs [35]. This method is generally performed using organic solvents that are partially miscible with water, such as methyl acetate, ethyl acetate, isopropyl acetate, benzyl alcohol, and butyl lactate. Initially, the organic solvent and water are mutually saturated with each other to obtain the initial thermodynamic equilibrium of both phases. Lipids and drugs are dissolved in the water-saturated solvent, which is then emulsified in the aqueous phase (solvent-saturated water-containing stabilizer) under stirring to form an o/w emulsion. The emulsion is diluted with water (volume ratio from 1:5 to 1:10) to allow diffusion of the solvent into the continuous phase. SLNs and NLCs are formed spontaneously due to lipid precipitation and the solvent is then eliminated by lyophilization or vacuum distillation [36]. This method can avoid exposing drugs to high temperatures and physical stress like those associated with high-speed stirring or high-pressure homogenization. In addition, it can be easily scaled up. Furthermore, the technique can be applied to hydrophilic and hydrophobic drugs. Different drugs and biomolecules, such as insulin, tretinoin, and cyclosporine have been encapsulated in SLNs and NLCs using this technique. However, like the microemulsion method, the solvent emulsification-diffusion method is associated with substantial dilution of SLNs and NLCs dispersions and requires a purification process to remove residual organic solvent [18].

2.5. Solvent Emulsification-Evaporation Method

Unlike the emulsification-diffusion method, water-immiscible organic solvents (e.g., chloroform, cyclohexane, dichloromethane, and toluene) are used to prepare SLNs and NLCs using the solvent emulsification-evaporation method [37]. Briefly, drugs and lipids are dissolved in a solvent or a solvent mix and then emulsified in an aqueous phase to form nanodispersions. Thereafter, the organic solvent is evaporated by mechanical stirring or in a rotary evaporator. SLNs and NLCs are formed due to lipid precipitation after solvent evaporation [38]. The concentration of lipids in the organic phase has a considerable effect on mean PSs of SLNs and NLCs prepared using the solvent emulsification-evaporation method. Small SLNs and NLCs are obtained at a low lipid concentration [1]. The main advantage of this method is avoidance of drug exposure to high temperatures, and thus, it is useful for encapsulating highly thermo-labile drugs. Furthermore, SLNs and NLCs produced using this method and the solvent emulsification-diffusion method have PSs of around 100 nm and narrow size distributions. The limitations of the solvent emulsification-evaporation method are that it requires toxic organic solvents and the suspension produced is dilute and requires further evaporation or ultra-filtration [29]. Both the solvent emulsification-diffusion and solvent emulsification-evaporation methods involve the use of organic solvents, which means additional solvent removal steps are needed. Furthermore, in vitro and in vivo risk assessments on the SLNs and NLCs produced are also required. The residual solvent levels should be below specified values, at which no adverse effect is observed [39].

2.6. Double Emulsion Method

The double emulsion method provides a means of producing SLNs and NLCs of hydrophilic drugs and biomolecules (e.g., peptides and proteins) [40]. Particularly, it has been widely used to incorporate insulin into the matrices of SLNs and NLCs for oral delivery [41]. According to this method, a drug and a stabilizer are dissolved in an aqueous solution and then emulsified in a water-immiscible organic phase containing lipids [42] or in solvent-free molten lipids [43][44]. These primary emulsions are dispersed in an aqueous phase containing a hydrophilic emulsifier to form water/oil/water (w/o/w) emulsion. After solvent evaporation, SLNs and NLCs dispersions are obtained due to lipid precipitation [45]. Notably, for this method, the stabilizer essentially prevents drug partitioning to the external water phase during the solvent evaporation. The advantage of the double emulsion method is that it does not require molten lipid. However, EE and DL values of SLNs and NLCs are low due to the leaching of the hydrophilic drugs to the outer aqueous phase. EEs of SLNs and NLCs produced using this technique may range from 5.2–40.3% for insulin [45] or relatively higher (63%) for sulforhodamine 101 [44]. In some cases, EEs can also reach 80% for diethyldithiocarbamate [43] and 90% for polymyxin B sulfate [46]. In addition, the SLNs and NLCs prepared using this method have relatively large PSs (up to micro sizes) [18].

2.7. Phase Inversion Temperature (PIT) Method

The PIT method has been used to produce nanoemulsions, SLNs, and NLCs [47]. The technique is based on the temperature-induced inversions of w/o to o/w emulsions and vice versa. The method requires the use of non-ionic polyoxyethylated surfactants with temperature-dependent properties. The hydrophilic-lipophilic balance (HLB) value of these surfactants is high at low temperatures because their hydrophilic groups are highly hydrated. When the temperature is increased, dehydration of the ethoxy group occurs, which increases surfactant lipophilicity and decreases HLB values of the surfactants [48]. PIT is defined as the temperature at which the affinities of the surfactants for aqueous and lipid phases are equal [49]. At temperatures > PIT, the surfactants favor the formation of w/o emulsions, whereas, at temperatures < PIT, they turn to form o/w emulsions [50]. To produce SLNs and NLCs, oil, water, and surfactant are first heated to a temperature > PIT under stirring to form w/o emulsions. Subsequently, they are rapidly cooled with continuous stirring, which promotes the breakdown of w/o microemulsions and induces the formation of o/w nanoemulsions. SLNs and NLCs are formed when lipids are precipitated at low temperatures [51]. SLNs and NLCs prepared using this method have been reported to have small PSs, narrow size distribution, and excellent stabilities [52]. This method requires little energy input and does not require organic solvents. However, it involves low stabilities of the nanoemulsion formed, and sometimes requires several temperature cycles (e.g., three cycles between 60 and 90 °C) [53].

2.8. Membrane Contactor Method

In this method, a specific membrane contactor is used for producing SLNs and NLCs. Briefly, a lipid phase is pressed through the pores of a membrane while the temperature is maintained above the solid lipid’s melting point. This step results in the formation of small droplets. At the same time, an aqueous phase containing surfactants circulates over the other side of the membrane inside a module. It flows tangentially to the membrane surface and sweeps away droplets formed at pore outlets. SLNs and NLCs are formed by allowing the hot emulsion to cool down to room temperature [54]. The effects of formulation and process parameters, including the membrane pore size, lipid phase pressure, flow velocity of the aqueous phase, and temperatures of the aqueous and lipid phases on SLNs and NLCs size are intensively investigated. PS can be adjusted by changing lipid phase flux through membranes. This method has been used to prepare vitamin E-loaded liposomes, micelles, nanoemulsions, and SLNs for pulmonary drug delivery [55]. This method is feasible for scaling-up as a membrane with a pore size of 0.1 μm resulted in high fluxes [56]. However, the limitations of this method are that it requires a sophisticated system and membranes are prone to clogging.

2.9. Supercritical Fluid-Based Methods

Several SLNs and NLCs production methods involved the use of supercritical fluids like supercritical CO2. For the supercritical fluid extraction of emulsions (SFEE) method, an o/w emulsion is prepared beforehand, followed by supercritical fluid extraction of the organic solvent. Typically, the emulsion is added to an extraction column from the top, and supercritical CO2 is introduced in a counter-current manner from the bottom. SFEE has a much higher solvent extraction efficiency than other methods that use evaporation, diffusion, and dilution. The solvent is quickly and completely removed, and this leads to lipid precipitation. Furthermore, the produced SLNs and NLCs have uniform particle size distribution [57]. The o/w emulsions are prepared using water-immiscible solvents [57] and water-partially miscible solvents [58]

Supercritical CO2 is also used in a method called supercritical assisted injection in a liquid antisolvent (SAILA). This method is similar to the solvent injection method, which is discussed below. In brief, lipids and drugs are dissolved in a water-miscible organic solvent, mixed with supercritical CO2 and injected into an aqueous phase containing surfactant. The mixing of the two fluids results in a rapid supersaturation and consequent precipitation of SLNs or NLCs [59]. Supercritical fluid-based methods usually result in uniform particle size distributions and their solvent extraction efficiencies are higher than those of conventional extraction methods. However, they are expensive and involve the use of organic solvents [58].

2.10. Coacervation Method

Coacervation has been widely used to produce polymeric nanoparticles and was first used by Battaglia et al. to prepare SLNs in 2010 [60]. This method is based on the acidification-induced precipitation of the alkaline salts of fatty acids. The lipids used are in alkaline salt forms (e.g., sodium stearate), which are dispersed in an aqueous solution of a polymeric stabilizer, such as PVA or HMPC [61]. Drugs can be dissolved in the lipid phase [62] or loaded into blank SLNs in later steps [63]. To load drugs into the lipid phase, drugs are solubilized in ethanol and dissolved in the lipid. The mixture is heated to a temperature above its Krafft point when a clear micellar solution of the lipid alkaline salts is formed. An acidifying solution (coacervating solution) is then added dropwise to this solution, which causes the lipids to precipitate. The obtained suspension is then cooled in a water bath with stirring at 300 rpm until a temperature of 15 °C is reached then cooled in a water bath with stirring to complete the precipitation of SLNs or NLCs [64]. The coacervation method provides a simple solvent-free means of preparing SLNs and NLCs without sophisticated instruments. However, it can only be used on lipids that form alkaline salts, such as fatty acids, and is not suitable for pH-sensitive drugs [64].

2.11. Solvent Injection Method

The solvent injection method used for preparing SLNs and NLCs was first reported by Schubert et al. in 2003 [65]. According to this method, lipids and drugs are dissolved in a water-miscible solvent (e.g., methanol, ethanol, isopropanol, or acetone) or a water-miscible solvent mixture. The aqueous phase is usually prepared by adding an emulsifier or an emulsifier mixture to water or a buffer solution. The organic phase is then quickly injected into the aqueous phase under continuous mechanical stirring using a needle [18]. The basic principles of this method and the solvent emulsification-diffusion method are similar. Following injection, two principal mechanisms occur simultaneously and aid each other to form SLNs and NLC. First, the solvent diffuses out of the droplets into the aqueous phase, which results in a droplet size reduction. As a consequence, lipid concentration within the droplets increases, which leads to the formation of local supersaturated regions stabilized by emulsifiers in the aqueous phase [65]. Second, the emulsifiers reduce the interfacial tension between water and solvent, and this leads to the formation of small solvent-lipid droplets at the injection site. Due to the interfacial pulsation and turbulence during solvent diffusion, those droplets are broken into smaller droplets with essentially the same lipid concentrations [65]. The free energy released when the solvent is redistributed to its equilibrium state provides the energy required for droplet division [65]. Therefore, in the solvent injection method, solvent diffusion results in the formation of tiny droplets and lipid precipitation. Emulsifiers play an important role in the determination of PSs and size distributions. Consequently, SLNs and NLCs are formed and stabilized by the emulsifier. Notably, the diffusion rate of organic solvent into the aqueous phase is considered to be one of the most critical factors affecting PSs and size distributions [65][66].

This method has been modified in some studies, including a micro-channel with a cross-shaped junction [67] or a co-flowing micro-channel system [68]. The micro-channel with a cross-shaped junction consists of a microchannel module, two precision syringe pumps for supplying an aqueous phase and a solution of lipid in solvent, a digital inversion microscope, and a stirred unit to collect the SLNs suspension. The process was carried out as follows: an organic solution (Softisan 100 in acetone, viscosity 0.35 mPa) was fed into the main microchannel, and an aqueous phase (0.5% poloxamer 188, viscosity 1.46 mPa) was pumped into the inlets of the two branch channels at an equal flow rate. A digital inversion microscope system was used to capture images of flow field behaviors in microchannels. The produced SLNs had PSs of 100–200 nm [67]. In the co-flowing micro-channel system, two precision syringe pumps were also used to supply the aqueous and lipid-solvent phase. The inner capillary had inner and external diameters of 110 and 490 μm, respectively, whereas these values of the outer capillary were 650 and 7000 μm, respectively. An organic solution (Softisan 100 in acetone) was fed into the inner capillary, and simultaneously, an aqueous phase (poloxamer 188) was pumped into the outer capillary at the same flow rate. The produced SLNs had small PSs (<250 nm) and narrow size distribution (PDIs <0.26) [68]. Another modification of the solvent injection method is the microfluidic rapid ethanol dilution method using baffle devices. This method was used as a post-treatment and provided precise size control of SLNs and NLCs prepared using the solvent injection method. For the SLNs and NLCs preparation step, 1-palmitoyl-2-oleoyl-sn-glycero-3-phosphocholine (POPC) in ethanol and saline were introduced into the baffle device. Post-treatment was performed on-device and involved diluting the SLNs and NLCs suspensions (with 25% v/v ethanol) to 1%. This post-treatment was applied to prepare siRNA-loaded NLCs with a small PS (33 nm) and a high EE (>90%). The formulation specifically knocked down the plasma coagulation factor VII [69]. Table 1 summarizes the mechanisms, advantages, and disadvantages of different SLNs and NLCs preparation methods.

Table 1. Mechanisms, advantages, and disadvantages of different SLNs and NLCs preparation methods.

Method Mechanism Advantage Disadvantage
Hot high-pressure homogenization High shear stress and cavitational forces Speed, straightforward, avoidance of organic solvents, scalability Drug degradation under high temperature, drug loss into the aqueous phase
Cold high-pressure homogenization High shear stress and cavitational forces Prevention of drug degradation, applicability to hydrophilic drugs Large particles, broad size distributions
High-speed stirring and ultra-sonication High shear between two solid
adjacent area
Formation, growth, and implosive collapse
of bubbles in a liquid
straightforward, avoidance of organic solvents, low cost, scalability Exposure of drugs to high temperatures, metal contamination from sonicator probes, high surfactant concentrations, low lipid concentrations
Microemulsion Spontaneous interfacial
tension reduction under dilution
Simplicity, reproducibility, scalability, avoidance of organic solvents Large amount of water to dilute microemulsions, high concentration of surfactants
Solvent emulsification-diffusion Diffusion of solvent from lipid phase to aqueous phase leading to lipid precipitation Simplicity, avoidance of heat, small PS, narrow size distribution
Scalability, applicability to both hydrophilic and hydrophobic drugs
Residual solvent, additional solvent removal procedures
Solvent emulsification-evaporation method Evaporation of solvent in lipid phase leading to lipid precipitation Simplicity, avoidance of heat, small PS, narrow size distribution Residual solvent, additional solvent removal procedure
Dilute suspensions, requirement of evaporation or ultra-filtration
Double emulsion Lipid crystallization due to solvent evaporation or low temperature Applicability to hydrophilic drugs Low EE and DL
Large PS
Phase inversion temperature (PIT) Spontaneous inversion between oil/water and water/oil emulsions
with temperature change
Low energy, avoidance of organic solvents, narrow size distribution, good stability Instability of emulsion
Membrane contactor Formation of small
droplets after pressing lipid phase through membrane pores
Scalability, control of size Clogging of membrane
Supercritical fluid-based methods Quick evaporation or diffusion of solvent with the help of supercritical fluid, resulting in lipid precipitation Uniform particle size distribution, high solvent extraction efficiency Use of organic solvent, high expense
Coacervation Precipitation of alkaline salts of fatty acids when decreasing pH Simplicity, no sophisticated instrument, avoidance of organic solvents Applicability only to lipids in alkaline salt form and non pH-sensitive drugs
Solvent injection Diffusion of solvent from lipid phase to aqueous phase leading to lipid precipitation Simplicity, straightforward, fast production process, no sophisticated instrument Residual solvent, additional solvent removal procedure

References

  1. Wolfgang Mehnert; Mäder, K.; Solid lipid nanoparticles Production, characterization and applications. Advanced Drug Delivery Reviews 2001, 47, 165-196, 10.1016/s0169-409x(01)00105-3.
  2. Müller, R.H.; Mäder, K.; Gohla, S.; Solid lipid nanoparticles (SLN) for controlled drug delivery: a review of the state of the art. European Journal of Pharmaceutics and Biopharmaceutics 2000, 50, 161-177, 10.1016/s0939-6411(00)00087-4.
  3. Slavomira Doktorovova; Eliana B. Souto; Amélia M. Silva; Nanotoxicology applied to solid lipid nanoparticles and nanostructured lipid carriers – A systematic review of in vitro data. European Journal of Pharmaceutics and Biopharmaceutics 2014, 87, 1-18, 10.1016/j.ejpb.2014.02.005.
  4. Jana Pardeike; Aiman Hommoss; Rainer H. Müller; Lipid nanoparticles (SLN, NLC) in cosmetic and pharmaceutical dermal products. International Journal of Pharmaceutics 2009, 366, 170-184, 10.1016/j.ijpharm.2008.10.003.
  5. Medha D. Joshi; Rainer H. Müller; Lipid nanoparticles for parenteral delivery of actives. European Journal of Pharmaceutics and Biopharmaceutics 2009, 71, 161-172, 10.1016/j.ejpb.2008.09.003.
  6. Kumar A. Shah; Abhijit A. Date; Medha D. Joshi; Vandana B. Patravale; Solid lipid nanoparticles (SLN) of tretinoin: Potential in topical delivery. International Journal of Pharmaceutics 2007, 345, 163-171, 10.1016/j.ijpharm.2007.05.061.
  7. Laura Ravani; Elisabetta Esposito; Christian Bories; Vanessa Liévin-Le Moal; Philippe M. Loiseau; Madeleine Djabourov; Rita Cortesi; Kawthar Bouchemal; Clotrimazole-loaded nanostructured lipid carrier hydrogels: Thermal analysis and in vitro studies. International Journal of Pharmaceutics 2013, 454, 695-702, 10.1016/j.ijpharm.2013.06.015.
  8. Elisabetta Esposito; Maddalena Sguizzato; Markus Drechsler; Paolo Mariani; Federica Carducci; Claudio Nastruzzi; Rita Cortesi; Progesterone lipid nanoparticles: Scaling up and in vivo human study. European Journal of Pharmaceutics and Biopharmaceutics 2017, 119, 437-446, 10.1016/j.ejpb.2017.07.015.
  9. Van-An Duong; Thi-Thao-Linh Nguyen; Han-Joo Maeng; Sang-Cheol Chi; Preparation of Ondansetron Hydrochloride-Loaded Nanostructured Lipid Carriers Using Solvent Injection Method for Enhancement of Pharmacokinetic Properties. Pharmaceutical Research 2019, 36, 138, 10.1007/s11095-019-2672-x.
  10. Maria Luisa Bondi'; Emanuela Fabiola Craparo; Solid lipid nanoparticles for applications in gene therapy: a review of the state of the art. Expert Opinion on Drug Delivery 2009, 7, 7-18, 10.1517/17425240903362410.
  11. Karl Hörmann; Andreas Zimmer; Drug delivery and drug targeting with parenteral lipid nanoemulsions — A review. Journal of Controlled Release 2016, 223, 85-98, 10.1016/j.jconrel.2015.12.016.
  12. Sergio Calva-Estrada; Oscar García; María Remedios Mendoza; Maribel Jiménez; Characterization of O/W emulsions of carotenes in blackberry juice performed by ultrasound and high-pressure homogenization. Journal of Dispersion Science and Technology 2017, 39, 181-189, 10.1080/01932691.2017.1306783.
  13. R. Shegokar; K.K. Singh; R.H. Müller; Production & stability of stavudine solid lipid nanoparticles—From lab to industrial scale. International Journal of Pharmaceutics 2011, 416, 461-470, 10.1016/j.ijpharm.2010.08.014.
  14. Caibiao Hu; Airui Qian; Qiang Wang; Feng Xu; Yi He; Jing Xu; Yongchang Xia; Qiang Xia; Industrialization of lipid nanoparticles: From laboratory-scale to large-scale production line. European Journal of Pharmaceutics and Biopharmaceutics 2016, 109, 206-213, 10.1016/j.ejpb.2016.10.018.
  15. Franz Suter; Daniel Schmid; Franziska Wandrey; Fred Zülli; Heptapeptide-loaded solid lipid nanoparticles for cosmetic anti-aging applications. European Journal of Pharmaceutics and Biopharmaceutics 2016, 108, 304-309, 10.1016/j.ejpb.2016.06.014.
  16. Jana Štecová; Wolfgang Mehnert; Tobias Blaschke; Burkhard Kleuser; Ramadurai Sivaramakrishnan; Christos C. Zouboulis; Holger Seltmann; Hans Christian Korting; Klaus D. Kramer; Monika Schäfer-Korting; et al. Cyproterone Acetate Loading to Lipid Nanoparticles for Topical Acne Treatment: Particle Characterisation and Skin Uptake. Pharmaceutical Research 2007, 24, 991-1000, 10.1007/s11095-006-9225-9.
  17. Shweta Gupta; Rajesh Kesarla; Narendra Chotai; Ambikanandan Misra; Abdelwahab Omri; Systematic Approach for the Formulation and Optimization of Solid Lipid Nanoparticles of Efavirenz by High Pressure Homogenization Using Design of Experiments for Brain Targeting and Enhanced Bioavailability. BioMed Research International 2017, 2017, 1-18, 10.1155/2017/5984014.
  18. Surajit Das; Anumita Chaudhury; Recent Advances in Lipid Nanoparticle Formulations with Solid Matrix for Oral Drug Delivery. AAPS PharmSciTech 2010, 12, 62-76, 10.1208/s12249-010-9563-0.
  19. Danielle Irby; Chengan Du; Feng Li; Lipid–Drug Conjugate for Enhancing Drug Delivery. Molecular Pharmaceutics 2017, 14, 1325-1338, 10.1021/acs.molpharmaceut.6b01027.
  20. Yongtao Duan; Abhishek Dhar; Chetan Patel; Mehul Khimani; Swarnali Neogi; Prolay Sharma; Nadavala Siva Kumar; Rohit L. Vekariya; A brief review on solid lipid nanoparticles: part and parcel of contemporary drug delivery systems. RSC Advances 2020, 10, 26777-26791, 10.1039/d0ra03491f.
  21. Van-An Duong; Thi-Thao-Linh Nguyen; Han-Joo Maeng; Sang-Cheol Chi; Nanostructured lipid carriers containing ondansetron hydrochloride by cold high-pressure homogenization method: Preparation, characterization, and pharmacokinetic evaluation. Journal of Drug Delivery Science and Technology 2019, 53, 101185, 10.1016/j.jddst.2019.101185.
  22. Van-An Duong; Thi-Thao-Linh Nguyen; Han-Joo Maeng; Sang-Cheol Chi; Data on optimization and drug release kinetics of nanostructured lipid carriers containing ondansetron hydrochloride prepared by cold high-pressure homogenization method. Data in Brief 2019, 26, 104475, 10.1016/j.dib.2019.104475.
  23. Yub Raj Neupane; M D Sabir; Nafees Ahmad; Mushir Ali; Kanchan Kohli; Lipid drug conjugate nanoparticle as a novel lipid nanocarrier for the oral delivery of decitabine: ex vivo gut permeation studies. Nanotechnology 2013, 24, 415102, 10.1088/0957-4484/24/41/415102.
  24. Eliana B. Souto; Slavomira Doktorovova; Aleksandra Zielinska; Amélia M. Silva; Doktorovova S.; Key production parameters for the development of solid lipid nanoparticles by high shear homogenization.. Pharmaceutical Development and Technology 2019, 24, 1181-1185, 10.1080/10837450.2019.1647235.
  25. Shuyu Xie; Luyan Zhu; Zhao Dong; Xiaofang Wang; Yan Wang; Xihe Li; Wenzhong Zhou; Preparation, characterization and pharmacokinetics of enrofloxacin-loaded solid lipid nanoparticles: Influences of fatty acids. Colloids and Surfaces B: Biointerfaces 2011, 83, 382-387, 10.1016/j.colsurfb.2010.12.014.
  26. Fardin Tamjidi; Mohammad Shahedi; Jaleh Varshosaz; Ali Nasirpour; Design and characterization of astaxanthin-loaded nanostructured lipid carriers. Innovative Food Science & Emerging Technologies 2014, 26, 366-374, 10.1016/j.ifset.2014.06.012.
  27. Ali Akhoond Zardini; Mohebbat Mohebbi; Reza Farhoosh; Shadi Bolurian; Production and characterization of nanostructured lipid carriers and solid lipid nanoparticles containing lycopene for food fortification. Journal of Food Science and Technology 2017, 55, 287-298, 10.1007/s13197-017-2937-5.
  28. Vincent Jannin; Lucia Blas; Stéphanie Chevrier; Cédric Miolane; Frédéric Demarne; Denis Spitzer; Evaluation of the digestibility of solid lipid nanoparticles of glyceryl dibehenate produced by two techniques: Ultrasonication and spray-flash evaporation. European Journal of Pharmaceutical Sciences 2018, 111, 91-95, 10.1016/j.ejps.2017.09.049.
  29. Rita Cortesi; Production of lipospheres as carriers for bioactive compounds. Biomaterials 2002, 23, 2283-2294, 10.1016/s0142-9612(01)00362-3.
  30. S.A Wissing; O Kayser; R.H Müller; Solid lipid nanoparticles for parenteral drug delivery. Advanced Drug Delivery Reviews 2004, 56, 1257-1272, 10.1016/j.addr.2003.12.002.
  31. Rohan M. Shah; Daniel S. Eldridge; Enzo A. Palombo; Ian H. Harding; Microwave-assisted microemulsion technique for production of miconazole nitrate- and econazole nitrate-loaded solid lipid nanoparticles. European Journal of Pharmaceutics and Biopharmaceutics 2017, 117, 141-150, 10.1016/j.ejpb.2017.04.007.
  32. Wadzanayi L. Masiiwa; Louis L. Gadaga; Intestinal Permeability of Artesunate-Loaded Solid Lipid Nanoparticles Using the Everted Gut Method. Journal of Drug Delivery 2018, 2018, 1-9, 10.1155/2018/3021738.
  33. L.G. Souza; E.J. Silva; A.L.L. Martins; M.F. Mota; R.C. Braga; E.M. Lima; M.C. Valadares; S.F. Taveira; R.N. Marreto; Development of topotecan loaded lipid nanoparticles for chemical stabilization and prolonged release. European Journal of Pharmaceutics and Biopharmaceutics 2011, 79, 189-196, 10.1016/j.ejpb.2011.02.012.
  34. M Igartua; P Saulnier; B Heurtault; B Pech; J E Proust; J L Pedraz; J P Benoit; Development and characterization of solid lipid nanoparticles loaded with magnetite. International Journal of Pharmaceutics 2002, 233, 149-157, 10.1016/s0378-5173(01)00936-x.
  35. Michele Trotta; Francesca Debernardi; Otto Caputo; Preparation of solid lipid nanoparticles by a solvent emulsification–diffusion technique. International Journal of Pharmaceutics 2003, 257, 153-160, 10.1016/s0378-5173(03)00135-2.
  36. F.Q Hu; H Yuan; H.H Zhang; M Fang; Preparation of solid lipid nanoparticles with clobetasol propionate by a novel solvent diffusion method in aqueous system and physicochemical characterization. International Journal of Pharmaceutics 2002, 239, 121-128, 10.1016/s0378-5173(02)00081-9.
  37. Brita Sjöström; Alon Kaplun; Yeshayahu Talmon; Bernard Cabane; Structures of Nanoparticles Prepared from Oil-in-Water Emulsions. Pharmaceutical Research 1995, 12, 39-48, 10.1023/a:1016278302046.
  38. Deep Pooja; Hitesh Kulhari; Lakshmi Tunki; Srinivas Chinde; Madhusudana Kuncha; Paramjit Grover; Shyam Sunder Rachamalla; Ramakrishna Sistla; Nanomedicines for targeted delivery of etoposide to non-small cell lung cancer using transferrin functionalized nanoparticles. RSC Advances 2015, 5, 49122-49131, 10.1039/c5ra03316k.
  39. Aleksandra Zielińska; Beatriz Costa; Maria V. Ferreira; Diogo Miguéis; Jéssica M. S. Louros; Alessandra Durazzo; Massimo Lucarini; Piotr Eder; Marco V. Chaud; Margreet Morsink; et al.Niels WillemenPatrícia SeverinoAntonello SantiniEliana B. Souto Nanotoxicology and Nanosafety: Safety-By-Design and Testing at a Glance. International Journal of Environmental Research and Public Health 2020, 17, 4657, 10.3390/ijerph17134657.
  40. M Garcı́a-Fuentes; D Torres; M.J Alonso; Design of lipid nanoparticles for the oral delivery of hydrophilic macromolecules. Colloids and Surfaces B: Biointerfaces 2003, 27, 159-168, 10.1016/s0927-7765(02)00053-x.
  41. Pedro Fonte; Tiago Nogueira; Christiane Gehm; Domingos Ferreira; Bruno Sarmento; Chitosan-coated solid lipid nanoparticles enhance the oral absorption of insulin. Drug Delivery and Translational Research 2011, 1, 299-308, 10.1007/s13346-011-0023-5.
  42. Mohsen Nabi-Meibodi; Alireza Vatanara; Abdolhossein Rouholamini Najafabadi; Mohammad Reza Rouini; Vahid Ramezani; Kambiz Gilani; Seyed Mohammad Hossein Etemadzadeh; Kayhan Azadmanesh; The effective encapsulation of a hydrophobic lipid-insoluble drug in solid lipid nanoparticles using a modified double emulsion solvent evaporation method. Colloids and Surfaces B: Biointerfaces 2013, 112, 408-414, 10.1016/j.colsurfb.2013.06.013.
  43. Karin Luize Mazur; Paulo Emílio Feuser; Alexsandra Valério; Arthur Poester Cordeiro; Camila Indiani De Oliveira; João Paulo Assolini; Wander Rogério Pavanelli; Claudia Sayer; Pedro H. H. Araújo; Diethyldithiocarbamate loaded in beeswax-copaiba oil nanoparticles obtained by solventless double emulsion technique promote promastigote death in vitro. Colloids and Surfaces B: Biointerfaces 2019, 176, 507-512, 10.1016/j.colsurfb.2018.12.048.
  44. Luana Becker Peres; Laize Becker Peres; Pedro Henrique Hermes de Araújo; Claudia Sayer; Solid lipid nanoparticles for encapsulation of hydrophilic drugs by an organic solvent free double emulsion technique. Colloids and Surfaces B: Biointerfaces 2016, 140, 317-323, 10.1016/j.colsurfb.2015.12.033.
  45. Marina Gallarate; Michele Trotta; Luigi Battaglia; Daniela Chirio; Preparation of solid lipid nanoparticles from W/O/W emulsions: Preliminary studies on insulin encapsulation. Journal of Microencapsulation 2008, 26, 394-402, 10.1080/02652040802390156.
  46. Patrícia Severino; Elisânia F. Silveira; Kahynna Loureiro; Marco V. Chaud; Danilo Antonini; Marcelo Lancellotti; Victor Hugo Sarmento; Classius F. da Silva; Maria Helena A. Santana; Eliana B. Souto; et al. Antimicrobial activity of polymyxin-loaded solid lipid nanoparticles (PLX-SLN): Characterization of physicochemical properties and in vitro efficacy. European Journal of Pharmaceutical Sciences 2017, 106, 177-184, 10.1016/j.ejps.2017.05.063.
  47. P. Izquierdo; Jordi Esquena; Th. F. Tadros; C. Dederen; M. J. Garcia; N. Azemar; Conxita Solans; Formation and Stability of Nano-Emulsions Prepared Using the Phase Inversion Temperature Method. Langmuir 2002, 18, 26-30, 10.1021/la010808c.
  48. Béatrice Heurtault; Patrick Saulnier; Brigitte Pech; Jacques‐Emile Proust; Jean‐Pierre Benoit; A Novel Phase Inversion-Based Process for the Preparation of Lipid Nanocarriers. Pharmaceutical Research 2002, 19, 875-880, 10.1023/a:1016121319668.
  49. L. Montenegro; M.G. Sarpietro; S. Ottimo; G. Puglisi; Francesco Castelli; Differential scanning calorimetry studies on sunscreen loaded solid lipid nanoparticles prepared by the phase inversion temperature method. International Journal of Pharmaceutics 2011, 415, 301-306, 10.1016/j.ijpharm.2011.05.076.
  50. Hasan Ali; Sandeep Kumar Singh; Preparation and characterization of solid lipid nanoparticles of furosemide using quality by design. Particulate Science and Technology 2017, 36, 695-709, 10.1080/02726351.2017.1295293.
  51. Songran Gao; David Julian McClements; Formation and stability of solid lipid nanoparticles fabricated using phase inversion temperature method. Colloids and Surfaces A: Physicochemical and Engineering Aspects 2016, 499, 79-87, 10.1016/j.colsurfa.2016.03.065.
  52. Lucia Montenegro; Claudia Carbone; Gabriele Condorelli; Rossella Drago; Giovanni Puglisi; Effect of Oil Phase Lipophilicity on In Vitro Drug Release from O/W Microemulsions with Low Surfactant Content. Drug Development and Industrial Pharmacy 2006, 32, 539-548, 10.1080/03639040600599806.
  53. C. Carbone; B. Tomasello; Barbara Ruozi; Marcella Renis; G. Puglisi; Preparation and optimization of PIT solid lipid nanoparticles via statistical factorial design. European Journal of Medicinal Chemistry 2012, 49, 110-117, 10.1016/j.ejmech.2012.01.001.
  54. Catherine Charcosset; Assma El-Harati; Hatem Fessi; Preparation of solid lipid nanoparticles using a membrane contactor. Journal of Controlled Release 2005, 108, 112-120, 10.1016/j.jconrel.2005.07.023.
  55. A. Laouini; V. Andrieu; L. Vecellio; H. Fessi; C. Charcosset; Characterization of different vitamin E carriers intended for pulmonary drug delivery. International Journal of Pharmaceutics 2014, 471, 385-390, 10.1016/j.ijpharm.2014.05.062.
  56. N. Khayata; W. Abdelwahed; M.F. Chehna; C. Charcosset; H. Fessi; Preparation of vitamin E loaded nanocapsules by the nanoprecipitation method: From laboratory scale to large scale using a membrane contactor. International Journal of Pharmaceutics 2012, 423, 419-427, 10.1016/j.ijpharm.2011.12.016.
  57. P. Chattopadhyay; B.Y. Shekunov; D. Yim; D. Cipolla; B. Boyd; S. Farr; Production of solid lipid nanoparticle suspensions using supercritical fluid extraction of emulsions (SFEE) for pulmonary delivery using the AERx system☆. Advanced Drug Delivery Reviews 2007, 59, 444-453, 10.1016/j.addr.2007.04.010.
  58. Roberta Campardelli; Maxime Cherain; Claire Perfetti; Carlo Iorio; MariaRosa Scognamiglio; Ernesto Reverchon; Giovanna Della Porta; Lipid nanoparticles production by supercritical fluid assisted emulsion–diffusion. The Journal of Supercritical Fluids 2013, 82, 34-40, 10.1016/j.supflu.2013.05.020.
  59. P. Trucillo; R. Campardelli; Production of solid lipid nanoparticles with a supercritical fluid assisted process. The Journal of Supercritical Fluids 2019, 143, 16-23, 10.1016/j.supflu.2018.08.001.
  60. Luigi Battaglia; Marina Gallarate; Roberta Cavalli; Michele Trotta; Solid lipid nanoparticles produced through a coacervation method. Journal of Microencapsulation 2009, 27, 78-85, 10.3109/02652040903031279.
  61. Daniela Chirio; Marina Gallarate; Elena Peira; Luigi Battaglia; Elisabetta Muntoni; Chiara Riganti; Elena Biasibetti; Maria Teresa Capucchio; Alberto Valazza; Pierpaolo Panciani; et al.Michele LanotteLaura AnnovazziValentina CalderaMarta MellaiGaetano FiliceSilvia CoronaDavide Schiffer Positive-charged solid lipid nanoparticles as paclitaxel drug delivery system in glioblastoma treatment. European Journal of Pharmaceutics and Biopharmaceutics 2014, 88, 746-758, 10.1016/j.ejpb.2014.10.017.
  62. Luigi Battaglia; Elisabetta Muntoni; Daniela Chirio; Elena Peira; Laura Annovazzi; Davide Schiffer; Marta Mellai; Chiara Riganti; Iris Chiara Salaroglio; Michele Lanotte; et al.Pierpaolo PancianiMaria Teresa CapucchioAlberto ValazzaElena BiasibettiMarina Gallarate Solid lipid nanoparticles by coacervation loaded with a methotrexate prodrug: preliminary study for glioma treatment. Nanomedicine 2017, 12, 639-656, 10.2217/nnm-2016-0380.
  63. Nausicaa Clemente; Benedetta Ferrara; Casimiro Luca Gigliotti; Elena Boggio; Maria Teresa Capucchio; Elena Biasibetti; Davide Schiffer; Marta Mellai; Laura Annovazzi; Luigi Cangemi; et al.Elisabetta MuntoniGianluca MiglioUmberto DianzaniLuigi BattagliaChiara Dianzani Solid Lipid Nanoparticles Carrying Temozolomide for Melanoma Treatment. Preliminary In Vitro and In Vivo Studies. International Journal of Molecular Sciences 2018, 19, 255, 10.3390/ijms19020255.
  64. Luigi Battaglia; Marina Gallarate; Elena Peira; Daniela Chirio; Ilaria Solazzi; Susanna Marzia Adele Giordano; Casimiro Luca Gigliotti; Chiara Riganti; Chiara Dianzani; Bevacizumab loaded solid lipid nanoparticles prepared by the coacervation technique: preliminary in vitro studies. Nanotechnology 2015, 26, 255102, 10.1088/0957-4484/26/25/255102.
  65. M.A. Schubert; Solvent injection as a new approach for manufacturing lipid nanoparticles – evaluation of the method and process parameters. European Journal of Pharmaceutics and Biopharmaceutics 2003, 55, 125-131, 10.1016/s0939-6411(02)00130-3.
  66. Ting Wang; Ning Wang; Yingying Zhang; Wancui Shen; Xingmei Gao; Tiefu Li; Solvent injection-lyophilization of tert-butyl alcohol/water cosolvent systems for the preparation of drug-loaded solid lipid nanoparticles. Colloids and Surfaces B: Biointerfaces 2010, 79, 254-261, 10.1016/j.colsurfb.2010.04.005.
  67. Songhong Zhang; Junxian Yun; Shaochuan Shen; Zhuo Chen; Kejian Yao; Jizhong Chen; Bingbing Chen; Formation of solid lipid nanoparticles in a microchannel system with a cross-shaped junction. Chemical Engineering Science 2008, 63, 5600-5605, 10.1016/j.ces.2008.08.005.
  68. Song-Hong Zhang; Shao-Chuan Shen; Zhuo Chen; Jun-Xian Yun; Ke-Jian Yao; Bing-Bing Chen; Ji-Zhong Chen; Preparation of solid lipid nanoparticles in co-flowing microchannels. Chemical Engineering Journal 2008, 144, 324-328, 10.1016/j.cej.2008.07.024.
  69. Niko Kimura; Masatoshi Maeki; Yusuke Sato; Akihiko Ishida; Hirofumi Tani; Hideyoshi Harashima; Manabu Tokeshi; Development of a Microfluidic-Based Post-Treatment Process for Size-Controlled Lipid Nanoparticles and Application to siRNA Delivery. ACS Applied Materials & Interfaces 2020, 12, 34011-34020, 10.1021/acsami.0c05489.
More
Information
Contributor MDPI registered users' name will be linked to their SciProfiles pages. To register with us, please refer to https://encyclopedia.pub/register :
View Times: 1.3K
Revisions: 2 times (View History)
Update Date: 20 Apr 2021
1000/1000
Video Production Service