Você está na página 1de 20

Sustainable Chemistry and Pharmacy 6 (2017) 3756

Contents lists available at ScienceDirect

Sustainable Chemistry and Pharmacy


journal homepage: www.elsevier.com/locate/scp

Lipid nanoparticles: Dierent preparation techniques, characterization, MARK


hurdles, and strategies for the production of solid lipid nanoparticles and
nanostructured lipid carriers for oral drug delivery

Poovi Ganesana,b, Damodharan Narayanasamya,
a
Department of Pharmaceutics, College of Pharmacy, SRM University, Kattankulathur 603203, Tamil Nadu, India
b
Department of Pharmaceutics, College of Pharmacy, Mother Theresa Post Graduate and Research Institute of Health Sciences, (A Govt. of Puducherry Institution),
Puducherry 605006, India

A R T I C L E I N F O A B S T R A C T

Keywords: The bioavailability of an orally administered drug primarily depends on its solubility in the GIT and its per-
Solid lipid nanoparticle meability across cell membranes. Also, a drug in solution form is preferred for conducting pharmacological,
Nanostructured lipid carrier toxicological and pharmacokinetic studies during the drug development stage. Thus, poor water solubility not
Hydrophobic drug only limits a drugs biological application but also challenges its pharmaceutical development. The use of lipid
Bioavailability
nanoparticles (LNs) in pharmaceutical technology has been reported for several years due to its important in
green chemistry for several reasons specically for its biochemical as green materials and biochemical pro-
cesses as green processes that can be very environmentally friendly. Also, the physiological/physiologically
related lipids (GRAS) made LNs usually enhance the drug absorption in the GIT. Hence, the pathways for ab-
sorption, metabolism, and transportation are present in the body, which may contribute to a large extent to the
bio-fate of the lipidic carrier. Moreover, the LNs improves the mucosal adhesion and increases their GIT re-
sidence time. The LNs with a solid matrix are two types: solid lipid nanoparticle (SLN) and nanostructured lipid
carrier (NLC). Also, their hydrophobic core provides a suitable environment for entrapment of hydrophobic
drugs to improve its bioavailability. This review highlights and discusses the simple and easily scaled-up novel
SLN and NLC along with their dierent production techniques, hurdles, and strategies for the production of LNs,
characterization, lyophilization and drug release. Also, this review summarizes the research ndings reported by
the dierent researchers regarding the dierent method of preparation, excipients and their signicant ndings.

1. Introduction the case of polymeric nanoparticles, the in vivo degradation of the


polymer might cause toxic eects (Das and Chaudhury, 2011; Mller
In the last few decades, various drug-delivery technologies have et al., 1996; Kumar, 2000). The physiochemical diversity and bio-
emerged (Jaiswal et al., 2016), and an interesting part of this has been compatibility of lipids and their ability to enhance oral bioavailability
the development of nanoscale drug delivery devices (Jaiswal et al., of drugs have made lipid nanoparticles very attractive carriers for oral
2004, 2016). Many nano particulate systems based on biocompatible drug delivery (Das and Chaudhury, 2011). Moreover, the lipid-based
polymers, lipids and oils have come to the fore, which can be eciently formulations can positively inuence drug absorption in a number of
used to increase the oral bioavailability of drugs either by increasing ways including; increasing solubilization capacity, preventing drug
the drug permeability or by overcoming the rst-pass eect and/or P-gp precipitation on intestinal dilution, enhancing membrane permeability,
eux (Harde et al., 2011). Among these, lipid-based nanoparticles have inhibiting eux transporters, reducing CYP enzymes, enhancing chy-
the advantage of being the least toxic for in vivo applications, and lomicron production and lymphatic transport (O'driscoll and Grin,
dynamic progress has been made in the area of DNA/RNA and drug 2008; Porter et al., 2007; Gursoy and Benita, 2004).
delivery using lipid-based nano assemblies (Puri et al., 2009). The lipids Lipid nanoparticles with a solid matrix are two types: solid lipid
used to prepare lipid nanoparticles are usually physiological lipids nanoparticle (SLN) and nanostructured lipid carrier (NLC) (Das and
(biocompatible and biodegradable) (Kaur et al., 2015) with low acute Chaudhury, 2011). The solid lipid nanoparticles (SLN) was introduced
and chronic toxicity (Das and Chaudhury, 2011; Mller et al., 1996). In as the rst generation of lipid nanoparticles (Iqbal et al., 2012;


Corresponding author.
E-mail address: poovinano@gmail.com (D. Narayanasamy).

http://dx.doi.org/10.1016/j.scp.2017.07.002
Received 22 May 2017; Received in revised form 24 July 2017; Accepted 25 July 2017
2352-5541/ 2017 Elsevier B.V. All rights reserved.
P. Ganesan, D. Narayanasamy Sustainable Chemistry and Pharmacy 6 (2017) 3756

Basavaraj, 2012), whereas the nanostructured lipid carrier (NLC), often These lipidic materials may contain puried triglycerides, complex
referred to as the second generation of SLN (Puri et al., 2009). The main glyceride mixtures, or waxes that are solid at both room temperature
reasons for the development of SLNs are combinational advantages and human body temperature (Wissing and Mller, 2003; Wissing et al.,
from dierent carrier systems such as liposomes and polymeric NPs. 2004) and are stabilized by suitable surfactant(s) (Kumar et al., 2012;
Similar to liposomes and nanoemulsions, they are composed of phy- Wissing et al., 2004; Wissing and Mller, 2003). Actually, SLN is an
siologically accepted biocompatible excipients (lipids and fatty acids). alternative carrier system to traditional colloidal carriers such as
Also, identical to polymeric nanoparticles, their solid matrix can e- emulsions, liposomes, and polymeric micro- and nanoparticles (Kumar
ciently protect the loaded active pharmaceutical ingredients against et al., 2012) and are interesting lipid-based drug-delivery carriers for a
chemical degradation under harsh biological milieu and provide the number of reasons, including i) particle size is on the nano- to sub-
maximum exibilities in the modication of the release proles of the micron scale (501000 nm) after drug encapsulation; ii) they are
drug. Further, they can be produced at large industrial scale by high- composed of biocompatible and biodegradable components (i.e., phy-
pressure homogenization (Harde et al., 2011; Kayser et al., 2005; siological lipids or lipid molecules) and do not require the use of or-
Muller et al., 2000; Mller et al., 1997; Freitas and Mller, 1999). All ganic solvents for their assembly; and iii) the particle synthesis process
these constructive attributes make the SLNs an excellent carriers for the (e.g., high-pressure homogenization) can be performed at a lower cost
oral drug delivery (Harde et al., 2011) and also for the pharmaceutical and are easily scaled up (Mudshinge et al., 2011; Wang et al., 2015;
formulation production process. Puri et al., 2009). Therefore, these nanoparticles bear the positive as-
Because pharmaceutical manufacturing is the most solvent-in- pects of other nano-lipid carrier systems, and they also overcome sev-
tensive and the least ecient of all chemical industries regarding waste eral of their disadvantages. For example, SLN is similar in nature to
generated per unit of product. Statistics compiled across the industry nanoemulsions but feature a solid lipid core as opposed to a liquid lipid
point to an average waste-to-product ratio of 200. In other words, version. As a result, drug mobility decreases in the solid lipid state
factories generate 200 kg of waste for every kilogram of active phar- compared with the oily phase, thereby enhancing the controlled release
maceutical ingredient produced and the nancial burden associated of loaded drugs. SLN stability can be further improved by the addition
with the processing and disposal of these sizeable waste-streams is of a surfactant coating (Mller et al., 2002; Pardeshi et al., 2012;
considerable (Rajagopal, 2014). Furthermore, pharmaceutical manu- Martins et al., 2007). An additional benet involves the production of
facturing plants devote exorbitant amounts of money each year for the SLN in a powder form, which may be loaded into pellets, capsules, or
fuel and electricity they need to keep their facilities running (Galitsky tablets for further development of drug delivery (Poovi, 2016; Puri
et al., 2008). As a counter to this, various green approaches have et al., 2009).
become popular as a means to reduce the ecological impact of the Despite these advantages, SLNs suer from few limitations, such as
pharmaceutical industry including the use of solvent-free synthetic low loading efciency (Pathak and Raghuvanshi, 2015), drug expulsion
procedures and alternative energy sources (Markarian, 2016). after polymorphic transition during storage, and relatively high water
The eld of green and sustainable chemistry has rapidly grown in content of the dispersions (7099.9%) (Kumar et al., 2012; Schwarz
the past few decades. Both in science and research as well as in seg- et al., 1994; Westesen et al., 1997). The poor drug loading capacity of
ments of the industry there have been many independent activities. conventional SLN is due to a densely packed lipid crystal network,
(Cannon and Warner, 2011; Kmmerer and Hempel, 2010; Angrick which allows little room for incorporation of drugs (Kumar et al., 2012)
et al., 2006) Green chemistry is a prerequisite and a core part of sus- or by the solubility of drug in the lipid melt, miscibility of drug melt and
tainable chemistry. Most important for green chemistry are the 12 lipid melt, the polymorphic state of the lipid matrix, and structure of
Principles of Green Chemistry (Anastas and Warner, 1998; Voutchkova- the solid matrix lipid (Schwarz et al., 1994, 2007).
Kostal et al., 2012). Green chemistry and sustainable chemistry have in To overcome the negative feature of SLN, a system of NLCs was
common a benign design, manufacture, and use of chemical products produced. The NLCs is composed of a mixture of spatially dierent lipid
and services which are ecient, eective, safe and environmentally molecules, normally the mixture of liquid lipid and solid, which makes
harmless. However, whereas green chemistry strives primarily at en- more imperfection in the matrix to incorporate more drug molecules
gineering and technical issues, like synthesis, atom economy and use of than solid lipid nanoparticles. Despite the presence of liquid lipid, NLC
solvents, and thereby derives from a perspective of design and pro- matrix is solid at body/room temperature. NLCs adopt mixtures of a
duction, sustainable chemistry encompasses all life cycle stages as well liquid lipid and solid lipid and remain in the solid state by controlling
as direct and indirect implications in surrounding areas and addresses the content of liquid lipid (Kaur et al., 2015). NLCs can more strongly
dierent perspectives besides environmental aspects like economy and immobilize drugs and prevent the particle from coalescing by the solid
society. matrix compared to emulsions. NLCs also have the advantages of SLNs
In this sense, the avoidance of organic solvents in the production including low toxicity, biodegradation, drug protection, slow release
process of SLNs and NLCs and its physiological lipid component makes and avoidance of organic solvents in production (Liu and Wu, 2010; Das
this system as a one of the challenging approaches among the for- and Chaudhury, 2011).
mulation scientist. Also, it complies the key objectives of Green In SLNs, the drug is mainly dispersed in molecular form, for ex-
chemistry and sustainable chemistry. This review highlights and dis- ample, located in between the fatty acid chains of the glycerides
cusses the two types of simple and easily scaled-up novel lipid nano- whereas in NLCs, a blend of solid and liquid lipids are used, and due to
particles with solid matrix (Solid lipid nanoparticles and their dierences in the structure they cannot t together very well to
Nanostructured lipid carriers) along with their dierent production form a perfect crystal. This arrangement creates a lot of imperfections
techniques, hurdles and strategies for the production of lipid nano- in matrix leading to an accommodation of more drugs in molecular
particles, characterization, lyophilization and drug release. Also, this form and amorphous clusters (Domingo and Saurina, 2012; Pardeshi
review summarizes the research ndings reported by the dierent re- et al., 2012).
searchers regarding the dierent method of preparation, excipients and
their signicant ndings.
3. Types of SLN/NLC
2. Solid lipid nanoparticles
Based on the chemical nature of the active ingredient and lipid,
Solid lipid nanoparticles are nanosphere composed of a solid lipids nature and concentration of surfactants, the solubility of the drug in the
core with a mean photon correlation spectroscopy (PCS) diameter be- melted lipid, type of production and the production temperature the
tween approximately 50 and 1000 nm (Battaglia and Gallarate, 2012). SLNs and NLCs are classied into three types (Table 1) (Figs. 1 and 2).

38
P. Ganesan, D. Narayanasamy Sustainable Chemistry and Pharmacy 6 (2017) 3756

Table 1
Description of SLNs and NLCs.

Type Nature of matrix Descriptions

Types of SLNs (Mehnert and Mder, 2001; Souto and Muller, 2007, 2010, 2006; Souto et al., 2004; Westesen et al., 1997; Muller et al., 2000)
Type I Homogeneous matrix model The SLN Type I is dened as the homogeneous matrix model because the API is molecularly dispersed in the lipid core or is
present in the form of amorphous clusters. This model is obtained when using optimized ratios of API and lipid passing
through the HPH at above the melting point of the lipid, or when using the cold HPH technique. As consequence of their
structure, SLN Type I can show controlled release properties.
Type II Drug enriched shell model It is obtained when the API concentration in the melted lipid is low. After applying the hot HPH technique, during the cooling
of the homogenized nanoemulsion, the lipid phase precipitates rst, leading to steadily increasing the concentration of API in
the remaining lipid melt with increased fraction of solidied lipid. An API free lipid core is formed; when the API reaches its
saturation solubility in the remaining melt, an outer shell containing both API and lipid will solidify around this core which
contains a low amount of API. This model is not suitable for prolonged API release; nevertheless, it may be used to obtain a
burst release of API, in addition to the occlusive properties of the lipid core.
Type III Drug enriched core model A drug enriched core model is obtained when the recrystallization mechanism is the opposite of that described for the drug
enriched shell model. In this method, a drug is solubilized in the lipid melt close to its saturation solubility. Subsequent
cooling of the lipid emulsion causes super-saturation of the drug in the lipid melt; this leads to the drug recrystallizing before
lipid recrystallization. Additional cooling leads to lipid recrystallization that forms a membrane around the already
crystallized drug-enriched core. This structural model is suitable for drugs that require prolonged release over a period.
Types of NLCs (Mller et al., 2002; Schfer-Korting, 2010; Shah et al., 2015; Jenning et al., 2000)
Imperfect Imperfectly structured solid matrix It is termed the imperfect crystal model and consists of a matrix with many voids and vacancies that can accommodate the
API. These particles are obtained when mixing solid lipids with a sucient amount of liquid lipids (oils). Because of the
dierent chain length of the fatty acid and the mixture of mono-, di- and triacylglycerols, the matrix of NLC is not able to form
a highly ordered structure, thus creating available spaces (structural imperfections).
Amorphous Structureless solid amorphous It is obtained when mixing special lipids (hydroxyl octacosanyl hydroxy stearate, isopropyl myristate, dibutyl adipate) that do
matrix not recrystallize after homogenization and cooling of the nanoemulsion (Gaba et al., 2015). These lipids create amorphous
matrices and during shelf life, thus minimizing API expulsion during storage time.
Multiple Multiple oils in fat in water The solubility of lipophilic the drug in liquid lipids (oils) is higher than in solid lipids. This principle can be used to develop
the multiple type NLC. In this type of NLC, a higher amount of oil are blended in solid lipids. At low concentrations, oil
molecules are easily dispersed into the lipid matrix. Additional of oil in excess of its solubility leads to phase separation
producing tiny oily nano-compartments surrounded by the solid lipid matrix. Such models allow controlled drug release, and
the lipid matrix prevents drug leakage. Lipophilic drugs can be solubilized in oils, and multiple types of NLCs are formed
during the cooling process of a hot homogenization process.

hazard generation ease of applicability and feasibility, and higher yield


potential (Malik et al., 2014). Major inroads have been made by
methods like ultra-sonication (Malik et al., 2014), Supercritical uid
technique (Konwarh et al., 2012), high-pressure homogenization
(Bevilacqua et al., 2007), ultraltration (Medina-Gonzlez et al., 2011),
occulation with surfactants (Malik et al., 2014), and many others
(Malik et al., 2014). All these green methods are most commonly used
method for the formulation of lipid nanoparticles (SLNs/NLCs)
The dierent approaches used for the production of SLNs/NLCs are
lavishly discussed and tabulated along with its advantages, dis-
advantages, mechanism, and limitations in Table 2. As well as, the
Fig. 1. Types of SLNs.
hurdles and strategies for the manufacturing SLNs and various SLNs/
NLCs formulations studied by dierent researchers to improve the oral
bioavailability of drugs and are also discussed in Tables 3 and 4 re-
spectively.

4.1. High energy approaches

4.1.1. High-Pressure Homogenization (HPH) technique


HPH is a reliable, well-established and powerful technique for the
large scale production. HPH has been used for years for the production
of nanoemulsions for parenteral nutrition (Lippacher et al., 2000;
Fig. 2. Types of NLCs. Mehnert and Mder, 2001). Recently, it has been applied for SLN and
NLC production and represents the main method established for these
4. Methods of preparation nanoparticles (Battaglia and Gallarate, 2012). In this technique, High-
pressure homogenizers push a liquid with high pressure (1002000 bar)
A number of interesting chemical approaches have been developed through a micron size gap: by applying high pressure, the liquid ac-
for the synthesis of nanoparticles. The most signicant and challenging celerates to high velocity (over 1000 km/h), and the resulting shear
parts concerned with the green method selection are the optimization of stress and cavitation forces break down the accelerated particles to
energy needs of the synthesis method and the corresponding energy submicron size (Mehnert and Mder, 2001). It can be performed either
constraints of the process. A number of traditional physical methods at elevated temperature (hot homogenization) or below room tem-
with reasonable modication in their methodology have been mastered perature (cold homogenization) (Schwarz et al., 1994). In both cases,
for controlled synthesis of nanoparticles. The distinguishing benets of the drug is dissolved or dispersed in the melted lipid at approximately
these methods range from their energy requirements, far less degree of 510 C above its melting point (All Rights are Reserved by Mrs and
Gupta, 2009; Emeje et al., 2012).

39
P. Ganesan, D. Narayanasamy Sustainable Chemistry and Pharmacy 6 (2017) 3756

Table 2
Comparison of various processes used for the preparation of SLNs/NLCs.

Comparison of various processes used for preparation of SLNs/NLCs

Method High-Pressure Homogenization (HPH) (Jorgensen and Nielson, 2009; Yadav et al., 2013; Yang et al., 1999; Mller et al., 2006; Almeida et al.,
1997; Harde et al., 2011; Mishra et al., 2012; Mudshinge et al., 2011)
Mechanism Shear due to intense turbulent eddies
Advantages Very eective dispersing technique
Disadvantages Extremely energy intensive process
Limitations for hot and cold Complete evasion of drug exposure to high temperature is not possible. Not appropriate for thermolabile drug (Iqbal et al., 2012). High
HPH polydispersity

Hot HPH (Jorgensen and Nielson, 2009; Yadav et al., 2013; Yang et al., 1999; Mller et al., 2006; Almeida et al., 1997; Harde et al., 2011; Mishra
et al., 2012; Mudshinge et al., 2011)
Mechanism Intense cavitations because of the large pressure drop through the valve
Advantages Scalable, commercially available
Disadvantages Drug distribution into the water phase during homogenization, Temperature-induced drug degradation (Swathi and Bala, 2013), Complexity of
crystallization step of nanoemulsion leading to several modications and supercooled melts

Cold HPH (Jorgensen and Nielson, 2009; Yadav et al., 2013; Yang et al., 1999; Mller et al., 2006; Almeida et al., 1997; Harde et al., 2011;
Mishra et al., 2012; Mudshinge et al., 2011)
Advantages No temperature induced drug degradation or crystalline modication
Disadvantages Not reported

Method Microemulsion Technique (Zara et al., 2002; Cavalli et al., 2003; Gasco, 1993; Harde et al., 2011; Mudshinge et al., 2011; Mishra et al., 2012;
Yadav et al., 2013)
Mechanism Spontaneous interfacial tension reduction
Advantages Low energy input, Theoretical stability
Disadvantages Extremely sensitive to change, Labor intensive formulation work, Low nanoparticle conc.
Limitations Strong dilution of particle suspension due to the use of the large volume of water.
High concentration of surfactant and co-surfactant is not desired.

Method Solvent Emulsication Evaporation Method (Conlin et al., 2010; Kang et al., 2010; Harde et al., 2011; Mudshinge et al., 2011; Yadav et al.,
2013; Sjstrm and Bergensthl, 1992)
Mechanism Emulsication (or diusion) of globules followed by evaporation leads to precipitation as particles
Advantages Small particle size 24 nm, Avoidance of heat, The low viscous system formed, Low energy input,
Lipids are dissolved in an aqueous immiscible solvent, e.g., cyclohexane, chloroform, Appropriate for thermolabile drugs (Iqbal et al.,
2012).
Disadvantages Low dispersing degree, Instability of emulsion, Insolubility of lipids in organic solvents, Additional solvent removal procedure, Toxicological
issue (residual solvent)
Limitations Production of very dilute nanoparticle dispersion is not required (Iqbal et al., 2012), An additional step is required. e.g. ultraltration or
evaporation, The organic solvent may remain in the nal preparation.

Method Solvent Emulsication Diusion Method (Conlin et al., 2010; Kang et al., 2010; Harde et al., 2011; Mudshinge et al., 2011; Yadav et al., 2013;
Trotta et al., 2003)
Mechanism Emulsication (or diusion) of globules followed by evaporation leads to precipitation as particles
Advantages Avoidance of heat during the production procedure, Lipids are dissolved in partially miscible solvent e.g. benzyl alcohol, Tetrahydrofuran.
Disadvantages Low dispersing degree, Instability of emulsion, Insolubility of lipids in organic solvents, Additional solvent removal procedure, Toxicological
issue (residual solvent)
Limitations Ultraltration or lyophilisation techniques are required, Residue of organic solvent may remain in the nal preparation.

Method Solvent Injection/Solvent Displacement Method (Schubert and Mller-Goymann, 2003; Iqbal et al., 2012)
Advantages Easy handling and fast production process, Lipids are dissolved in water missicible solvent. e.g. ethanol, methanol, acetone (Iqbal et al.,
2012) without using a sophisticated instrument (e.g., High-pressure homogenizer).

Method Membrane contractor (Ahmed El-Harati et al., 2006; Charcosset et al., 2005; Harde et al., 2011; Yadav et al., 2013)
Mechanism Lipid phase is forced through the membrane pores allowing the formation of small droplets (Iqbal et al., 2012)
Advantages Large-scale production, Facility of use, Control of size, In this case, the lipid phase is forced through the membrane pores to form small
droplets. Under cooling at room temperature (Iqbal et al., 2012), these droplets recrystallize forming the lipid nanoparticles.
Disadvantages Clogging of membrane

Method Phase Inversion Techniques (Montenegro et al., 2011a; Anton et al., 2008; Harde et al., 2011; Heurtault et al., 2002; Iqbal et al., 2012)
Mechanism Spontaneous inversion of o/w to w/o transitional emulsion with increase in temperature
Advantages Less energy intensive, Solvent free Good for heat liable molecules
Disadvantages Incorporation of additional molecules inuence inversion phenomenon
Instability of emulsion
Limitations Cumbersome technique

Method Coacervation Technique (Shah et al., 2015; Battaglia et al., 2010; Bianco et al., 2010; Chirio et al., 2011; Gallarate et al., 2010)
Mechanism Decrease in pH of micellar solution of alkaline salts of fatty acids by acidication (coacervating solution) in the presence of a polymeric stabilizer
causes proton exchange a lipid precipitation (coacervation)
Advantages Suitable for lipophilic drugs (by solubilising in the micellar solution after coacervation), Suitable for hydrophobic ion pairs of hydrophilic drugs,
Solvent-free technique, Use of sophisticated technique not required, Monodispersity, Simple to scale-up
Disadvantages Suitable for lipids that an form alkaline salts,
Not suitable for pH-sensitive drugs

Method Micro emulsion cooling technique


(Koziara et al., 2004, 2005; Mumper et al., 2006)
Mechanism Emulsication of globules followed by cooling leads to precipitation as particles
Advantages This method is reproducible, simple and easy to scale up; all the ingredients used are biocompatible; no organic solvents are used

Method Super Critical Fluid Method (Yadav et al., 2013; Chattopadhyay et al., 2006, 2007)
Mechanism Parallel processes of supercritical uid extraction (diusion) of organic solvent from emulsions and lipid dissolution;
Expansion of organic phase; leads to lipid crystallization
(continued on next page)

40
P. Ganesan, D. Narayanasamy Sustainable Chemistry and Pharmacy 6 (2017) 3756

Table 2 (continued)

Comparison of various processes used for preparation of SLNs/NLCs

Advantages Particles are obtained as a dry powder, Avoid the use of solvents; in preference to suspensions, Mild temperature and pressure conditions, Carbon
dioxide solution is the good choice as a solvent (Iqbal et al., 2012).
Disadvantages Very expensive method

Method Gas Assisted Melting Atomisation (Salmaso et al., 2009, Vezz et al., 2010)
Mechanism The lipid or protein/lipid mixtures were loaded into a thermostated mixing chamber (CM) where they were melted under supercritical CO2 at
selected temperature and pressure conditions. Then, the lipid saturated uid is forced through the nozzle by opening the valve at the bottom of
the CM, in order to produce microparticles.
Advantages Produce ne and non-agglomerated low density powders. Suitable operating conditions for protein-loaded lipid submicron particle preparation
were selected in order to obtain solid colloidal formulations and high protein loading while maintaining native activity and suitable release
proles.

Method Ultrasonication (Zhang et al., 2006; Xie et al., 2011; Mei et al., 2005; Harde et al., 2011)
Mechanism Formation and implosive collapse of bubbles due to cavitation, i.e., the formation, growth, and implosive collapse of bubbles in a liquid
Advantages Low particle size: 30180 nm, Low shear stress
Disadvantages Metal shading leads to contamination, Less entrapment eciency, Energy intensive process, Unproven scalability
Limitations During sonication, metallic contamination of the product may occur (Iqbal et al., 2012).

Mechanism of particle formation large extent, and it is required to obtain droplets in size range of a few
micrometers (Waghmare et al., 2012). In general, lower particle sizes
High mechanical shear due to strong turbulent eddies. was obtained as a result of higher temperatures due to the decreased
Lowering of pressure across the valves of homogenizers. viscosity of the inner phase (Lander et al., 2000). However, high tem-
Strong cavitation forces. peratures may also increase the degradation rate of the drug and the
carrier. In most of the cases, the homogenization step can be repeated
4.1.2. Hot homogenization technique several times. It should always be kept in mind, that high-pressure
In the hot homogenization method, the procedure is carried at homogenization increases the temperature of the sample (approxi-
temperatures above the melting temperature of the lipid (Svilenov and mately 108 C for 500 bar (Mehnert and Mder, 2001; Benita and Bhm,
Tzachev, 2014) (Fig. 3a). Here, the lipid and drug are melted and 1998). In most cases, 35 homogenization cycles at 5001500 bar are
combined with an aqueous surfactant at the same temperature. By using sufcient. Increasing the number of cycles or the homogenization
the high shear device, a hot pre-emulsion is formed. Usually, a Piston- pressure often results in an increase of the particle size (Waghmare
gap homogenizer or a jet-stream homogenizer is used to homogenize et al., 2012) due to particle coalescence which occurs as a result of the
the pre-emulsion to produce a hot colloidal emulsion. The droplets of high kinetic energy of the particles (Benita and Bhm, 1998; Mehnert
the hot colloidal emulsion are recrystallized by cooling the emulsion to and Mder, 2001). Then, the obtained nanoemulsion is cooled down to
room temperature in order to generate SLNs/NLCs. In some exceptional room temperature where the lipid recrystallizes and leads to the for-
cases, specic thermal treatment of emulsions such as cooling to re- mation of nanoparticles (Zur Mhlen et al., 1998).
frigeration conditions or even sub-zero temperatures may be required
(Bunjes et al., 1996; Lim and Kim, 2002; Schwarz and Mehnert, 1997; 4.1.3. Cold homogenization technique
Unruh et al., 2001). Cold homogenization has been developed to overcome the problems
The pre-emulsion quality aects the quality of the nal product to a of the hot homogenization technique (Jaiswal et al., 2016) such as a)

Table 3
Hurdles and strategies for the manufacturing of SLNs formulation (Mudshinge et al., 2011; Wu et al., 2011; Mishra et al., 2012) (Adapted and modied with permission from (Mishra
et al., 2012)).

Hurdles in SLN manufacture Strategies

1. High pressure-induced drug degradation High shear stress is the major cause of free radical formation and which results in decreased molecular weight of polymers
simultaneously causes:
Cavitation: can be solved by application of back pressure
As high molecular weight compounds are more sensitive to degradation, therefore low molecular weight compounds can be used

2. Lipid crystallization and drug incorporation Considerations:


The shape of lipid nanodispersions Platelet shapes have much larger surface areas compared to spheres; therefore, higher amount of the drug will be localized
directly on the surface of the particles
Gelation phenomena Gelation phenomena describes the transformation of a low-viscosity SLN dispersion into a viscous gel. It generally happens
during the i.v. injection into a living species, the life of this organism is put at risk. Gelation can be retarded or prevented by the
addition of co-emulsifying surfactants with high mobility (e.g. glycocholate); maintenance at optimum temperature range and
other environmental conditions
The presence of several lipid modications Lipid modications include improvement of quality like, lower density and ultimately, a higher capability to incorporate guest
molecules (e.g. drugs) which should be characterized by DSC, Xray, and NMR techniques
The existence of super cooled melts The main reason for the formation of supercooled melts is the size dependence of crystallization processes and thus provides
stability. The size of particles should be monitored maintained throughout the process
Co-existence of several colloidal species The presence of several colloidal species is an important point to consider as it may cause the degradation of the formulation. The
kinetics of the degradation will be determined by (1) the chemical reactivity of the drug, and (2) the concentration of the drug in
the aqueous medium or at the lipid/ water interface. By increasing the matrix, viscosity will decrease the diusion coecient of
the drug inside the carrier and therefore, will stabilize the formulation
Low drug loading capacity Improved by selection of proper drug carrier (nanosphere, nano pellets, etc.); use of the optimized drug: polymer ratio; selection
of suitable drug loading technique, etc.
Kinetics of distribution process The promoters of gelation (like high temperature, light, shear stress) increase the kinetic energy of the particles and favor
collision of the particles which produces instabilities to the formulations, therefore should be considered.

41
Table 4
Various research ndings of SLNs/NLCs formulations reported by dierent researches.

Type Drug Excipients Method Size (nm) Ref.

NLC Acitretin Precirol ATO 5, Labrasol Solvent diusion technique 223 8.92 nm Greater ecacy in the treatment of Psoriasis along with a reduction (Agrawal et al., 2010)
in side eects.
SLN All-trans retinoic acid Compritol 888 ATO, Soy HPH 150200 nm, 50100 nm Enhancement of GI absorption and improvement of oral (Hu et al., 2004b)
Lecithin/Pluronic F68 Or Soy bioavailability
P. Ganesan, D. Narayanasamy

Lecithin/Tween 80
NLC Alpha lipoic acid(LA) Apil, Miglyol 812, HPH 103 9 Suitable colloidal carrier for water insoluble LA and potential use for (Ruktanonchai et al.,
pluronic F68 topical delivery. 2009)
NLC Amphoteric B Theobroma Oil/Oleic Acid Solvent diusion method 479.67 11.56 nm Imparting stability to the NLC (Tan et al., 2010)
SLN Apomorphine Glyceryl monostearate, PEG, Probe soniation 63.20 0.98 nm to 174.63 3.19 nm Enhanced the bioavailability in rats (Tsai et al., 2011)
monostearate
SLN -Asarone Glyceryl caprate, Ultrasonic homogenization 199 nm Enhancement of GI absorption, Improvement of oral bioavailability, (Subedi et al., 2009)
Polyethylene glycol 660 tissue uptake and distribution
hydroxystearate
NLC Ascorbyl palmitate Imwitor 900, Labral, M1944 high pressure homogenization 170240 nm. Viscoelastic measurements is appropriate for topical/dermal (Teeranachaideekul
technique application et al., 2008)
SLN Baclofen Stearic Acid, Epikuron 200, Multiple (w/o/w) warm 161.4 nm Signicantly higher drug concentrations in plasma (Priano et al., 2011)
Propionic Acid, Butyric Acid, microemulsion
Sodium Taurocholate
NLC Benzocain Comprotol888 ATO, Ultrasonication 386.1 65.6 Targeting and prolonged release eects in dermal delivery. (Puglia et al., 2011)
Miglyol812, LutrolF68
SLN Buspirone HCl Cetyl Alcohol/Spermaceti, Emulsication-evaporation 86123nm Improvement of oral bioavailability (Varshosaz et al.,
Tween 20 or Poloxamer followed by ultrasonication 2010b)
NLC Calcipotriol Myverol 18-04K, Precirol Solvent evaporation method 267.3 12.3 Enhanced skin permeation, negligible skin irritation, and the (Lin et al., 2010)
Methtrexate ATO 5, Squalene, Pluronic compatibility of the two drugs.
F68

42
SLN Calcitonin Trimyristin w/o/w emulsion technique 200 nm Improvement of the eciency of such carriers for oral delivery of (Martins et al., 2009)
proteins
SLN Camptothecin Stearic acid, Soya lecithin, Hot HPH 196.8 21.3 Improved stability and sustained release eect (Yang et al., 1999)
Poloxamer 188
NLC Camptothecin Precirol ATO5, Compritol 888 HPH 192.3 10.2 Sustained drug release manner in decreasing order as follows (Huang, 2008)
ATO5, Myverol 18-04K, LE > NLC > SLN-P > SLN-C
Pluronic F68
NLC Camptothecin Monostearin, Soybean, Oil High pressure homogenize 117.3 2.8 nm Stable and high performance delivery system (Zhang et al., 2008)
788 and spray drying method. 216.6 4.1 nm
SLN Camptothecin Stearic Acid, Camptothecin, HPH 196.8 21.3 nm Sustained release and tissue targeting (Yang et al., 1999)
Soya Lecithin, Poloxamer
188, Glycerol
SLN Carvedilol Stearic Acid, Poloxamer 188, Microemulsion 120200 nm and 600800 nm Enhancement of oral bioavailability, Lymphatic uptake and bypass (Sanjula et al., 2009)
Sodium Taurocholate the hepatic rst-pass metabolism
SLN Clozapine Dynasan 114, Dynasan 116, Homogenization 96.7 163.3 Increased BA, high distribution to brain and reticuloendothelial cells (Manjunath et al.,
Dynasan 118, Epikuron 200, ultracentrifugation 2005)
Poloxamer 188
SLN Clozapine Trimyristin, tripalmitin, Hot homogenization, 96.7 3.8 to 163.3 0.7 nm Improvement of bioavailability (Manjunath and
tristearin ultrasonication method Venkateswarlu, 2005)
SLN Clozapine Trimyristin, Tripalmitin, Microemulsion method 87.2 46.9 nm Improvement of bioavailability (Xu et al., 2011)
Tristearin
SLN Clozapine Trimyristin, Tripalmitin, Hot homogenization followed 96.7 3.8 to 163.3 0.7 Improvement of oral bioavailability and tissue distribution (Manjunath and
Tristearin, Soylecithin 95%, by ultrasonication Venkateswarlu, 2005)
Poloxamer 188, Stearylamine
NLC Coenzyme Q 10 Miglyol812, Precifac ATO, HPH 195.9 3.6 Good physical stability and showed biphasic release pattern i.e. a fast (Obeidat et al., 2010)
Labrasol, Tegocare 450 release initially for skin saturation then slow and prolong release
prole to maintain the skin concentration of Q10.
(continued on next page)
Sustainable Chemistry and Pharmacy 6 (2017) 3756
Table 4 (continued)

Type Drug Excipients Method Size (nm) Ref.

SLN Crypto Tanshinone Glyceryl monostearate Ultrasonic and high-pressure 121.4 6.3 (GMS) and 137.5 7.1 (CP) Increases the solubilization capacity, changes the metabolism (Hu et al., 2010)
(GMS), Compritol 888 ATO homogenization method behavior and improved the oral bioavailability
(CP), Soya lecithin, Tween80,
Sodium dehydrocholate
SLN Crypto tanshinone Soy Lecithin, Tween 80, Ultrasonic and HPH 121.4 6.3 nm, 137.5 7.1 nm Enhancement of GI absorption and improvement of oral (Hu et al., 2010)
P. Ganesan, D. Narayanasamy

bioavailability.
NLC Curcumin GMS/MCT Nano-emulsication and between 120 and 250 nm Two fold increase in the anti-malarial activity (Nayak et al., 2010)
ultrasonication
SLN Curcumin Polysorbate 80, Soy Lecithin Microemulsion 134.6 nm Improvement of oral bioavailability and prolonged release (Kakkar et al., 2011)
SLN Cyclosporin A Glyceryl Monostearate, Melt- homogenization using a 131 nm and 158 nm Controlled release (Sawant et al., 2008)
Glyceryl Palmitostearate high-pressure homogenizer
SLN Cyclosporine A Imwitor, Tagat, sodium HPH 157 nm, 962 nm Low variation in bioavailability and avoidance of the huge initial (Mller et al., 2006)
cholate plasma concentration
SLN Cyclosporine A Imwitor 900, Tagat S, sodium Hot HPH 157 Improved bioavailability, less variation in plasma concentration (Mller et al., 2008)
cholate
NLC Cyproterone acetate Precirol, Oleic acid, Miglyol, HPH 200250 CPA incorporation in the NLC resulted in a 23 fold increase in (tecov et al., 2007)
poloxamer 188. CPA absorption.
SLN Dexamethasone Compritol 888 ATO Hot dispersion-ultrasonic 106.8 nm Drug delivery topical use (Chen et al., 2008)
technique
SLN Diazepam Compritol ATO 888, Imwitor Modied high-shear less than 500 nm Prolonged release (Abdelbary and
900 K homogenization and Fahmy, 2009)
ultrasound techniques
NLC Dihydroartemisnin GMS/Miglyol 812 Solvent diusion method 198 4.7 nm biphasic drug release pattern with burst release at the initial stage (Zhang et al., 2010)
and sustained release subsequently
NLC Docetaxel GMS, Soyabean lecithin, Ultrasonication dispersion 193.47 5.69 Sustained and continuous release pattern (Liu et al., 2011)
Stearic acid, Oleic acid,

43
Pluronic F68
NLC Domperidone Dynasan 114, Cetyl HPH 32.23 Fairly spherical shaped, a stable particle with controlled release. (Thatipamula et al.,
Resinoleate, Soy 2011)
Phosphatidylcholine, Tween
80
SLN Doxorubicin Glyceryl caprate Solvent emulsication- 199 nm Enhanced apoptotic death (Kang et al., 2010)
diusion method
NLC Etoposide GMS/soyabean oil Emulsication and low 125.991.2 nm Increased oral bioavailability and high cytotoxicity against human (Zhang et al., 2011)
temperature solidication epithelial- like lung carcinoma cells.
SLN Fenobrate Vitamin E TPGS, Vitamin E Hot HPH 58 Improved oral bioavailability (Hanafy et al., 2007)
6100
NLC Fluconazole Compritol 888 ATO (CA)/ Solvent diusion method 178 and 134 nm Good targeting eect along with sustained release and localized (Gupta and Vyas,
Oleic Acid eect 2012)
NLC Flurbiprofen (FB) Compritol 888 ATO, SA, HPH 179.7_3.102 highly eective, a non-irritant carrier for topical administration of (Gonzalez-Mira et al.,
Miglyol1 812, Castor Oil, FB and improved drug permeation 2010)
NLC Flurbiprofen Compritol ATO 888, Miglyol Probe ultrasonicator 55.4 Longer retention time due to mucoadhesive nature and improved (Liu et al., 2011)
812, Gelucire 44/14, Solutol penetration rate.
HS Tween 80 Glycerol
NLC Fluticasone Precirol ATO 5/labrasol Modied microemulsion Between 380 and 408 nm Improved the stability and loading capacity of the drug. (Doktorovov et al.,
Method 2010)
NLC Fluticasone Precirol ATO5, Labrasol, Microemulsion technique 316408 Particle size less than 1 m maintained over 60 days, and high EE (Doktorovov et al.,
propionate Tween80, Soyabean lecithin was achieved. 2010)
SLN Gonadotropin release Monostearin, Monoglyceride, Novel solvent diusion 421.7 nm Prolonged release (Hu et al., 2004a)
hormone chain method
SLN Hydrocortisone Length of the fatty acid Hot high pressure 151 4.2461 9.2 nm, Improved the stability and release properties of the drug (Jensen et al., 2010)
moiety homogenization
SLN Ibuprofen Stearic Acid, Triluarin, Solvent-free high-pressure 175189 nm Stable formulation and negligible cell cytotoxicity (Potta et al., 2011)
Tripalmitin homogenization (HPH)
SLN Idarubicin Stearic acid, Epikuron 200, Microemulsion 80 10 Improved BA, modies the PK and tissue distribution (Zara et al., 2002)
sodium taurocholate
Sustainable Chemistry and Pharmacy 6 (2017) 3756

(continued on next page)


Table 4 (continued)

Type Drug Excipients Method Size (nm) Ref.

SLN Idarubicin Emulsifying wax, Stearic acid, Microemulsion less than or around 100 nm Potential to deliver anticancer drugs (Ma et al., 2009)
octadecy alcohol, cetyl
palmitate,
NLC Indomethacine Compritol888ATO5, Ultrasonication 44.7191.8 High encapsulation eciency and delayed and sustained release (Ricci et al., 2005)
MiglyolATO5, LutrolF68, properties of the drug
P. Ganesan, D. Narayanasamy

Xanthum gum,
Carbopol934P
SLN Insulin Stearic acid, WGA-N-glut-PE, Ultrasonication 5860 Improved stability (Zhang et al., 2006)
Poloxamer 188, Soyalecithin
SLN Insulin Glyceryl Monostearate, Double emulsion method 216.8 30.9 nm Promising for oral delivery of proteins (Yang et al., 2011)
Glyceryl Palmitostearate,
Glyceryl Tripalmitate,
Glyceryl Behenate
SLN Insulin Cetyl Palmitate, Poloxamer Emulsion solvent diusion 320 26 nm and 361 30 nm Improvement of oral bioavailability (Sarmento et al., 2007)
407
NLC Itraconazole GMS, Precirol/Oleic Acid, Hot high-pressure 177 nm Stable NLCs were prepared which retained their properties during (Pardeike et al., 2011)
Miglyol homogenization nebulisation for pulmonary delivery.
NLC Ketoprofen Compritol888 ATO, Labrafac Ultrasonication 494 47 Improvement in the dissolution and skin permeation properties. (Cirri et al., 2012)
lipophile, Lutrol F68,
Xanthum gum
SLN Ketoprofen Mixture of beeswax and Microemulsion technique 75 4 nm to 250 9.38 nm Faster drug release (Kheradmandnia et al.,
carnauba wax 2010)
NLC Ketoprofen Compritol 888 ATO/ Labrafac Nanoemulsication and 300500 nm Improvement in both the dissolution and the skin permeation (Cirri et al., 2012)
Lipophile ultrasonication properties of drug
NLC Lidocaine Compritol888ATO, Ultrasound dispersion 72.1 Long duration of deep local anaesthesia in guinea pig. (Pathak and
PrecirolATO5, Nagarsenker, 2009b)

44
Miglyol812,T80
NLC Lidocaine Compritol 888 ATO/Miglyol Ultrasound dispersion method 78.1 nm Long duration of local anaesthesia in guinea pigs. (Pathak and
810 Nagarsenker, 2009a)
SLN Lopinavir Compritol 888 ATO, Pluronic Hot homogenization- 230 Avoid rst-pass metabolism (Alex et al., 2011)
F 127 ultrasonication
SLN Lovastatin Triglyceride, Hot homogenization- 60119 Avoid rst-pass metabolism and improved bioavailability (Suresh et al., 2007)
Phosphatidylcholine 95%, ultrasonication
poloxamer 188
NLC Lovastatin Squalene, Pluronic F68, Probe sonicator 180290 Controlled release of drug (Chen et al., 2010)
Precirol, ATO5, Myverol
1804k, Soyabean
phosphatidyl choline.
NLC Lutein Precirol ATO 5 Nano emulsication and 110 20 nm Lutein loaded NLC showed no release in simulated gastric uid and (Taratula et al., 2013)
ultrasonication slow release of lutein in simulated intestinal uid.
NLC Lutin Precirol ATO5, Myverol 18- Ultrasonic Emulsication 134 8 Lutein loaded NLC showed no release in simulated gastric uid and (Liu and Wu, 2010)
04k, CentrolN2F-SB, T20, slow release of lutein in simulated intestinal uid.
T80, Span 60 Pluronic F68,
Lauric, Palmitic, Myristic,
Stearic acid.
SLN Lysozyme/ peptides/ Witepsol E85, Softisan 142, Cold homogenization 540660 Improved stability, permeability and retention of integrity and (Rao, 2008)
vaccine cetyl alcohol, Poloxamer 182 activity
SLN Methotrexate Stearic acid, Monostearin, Solvent diusion method 120167 Improved oral bioavailability, enhancement of GI absorption and (Paliwal et al., 2009)
Tristearin, Compritol 888 lymphatic transport
ATO, L-a-soya lecithin
SLN N3-O-toluyl- ATO888, Soya Lecithin, Tfu Film 178.8 9.99 nm Enhancement of GI absorption and improvement of oral (Liu et al., 2010b)
uorouracil dispersionultrasonication bioavailability
SLN N3-O-toluyl- Lecithin, Compritol 888, Film 150200 nm Improvement of intestinal transport (Liu et al., 2010a)
uorouracil And Tfu dispersionultrasonication
(continued on next page)
Sustainable Chemistry and Pharmacy 6 (2017) 3756
Table 4 (continued)

Type Drug Excipients Method Size (nm) Ref.

NLC Nevirapine Steric Acid (SA), Oleic Acid Microemulsion 159.6 Uniform distribution of the particles and accelerated the release of (Kuo and Chung,
(OA), Compritol 888 ATO eective formulations in the delivery of drug for viral therapy. 2011)
Tween 80
SLN Nimesulide Palmitostearate, Glyceryl Hot homogenization process 230.4 5.6 nm Sustained drug release (Alex et al., 2011)
Tristearate
P. Ganesan, D. Narayanasamy

SLN Nitrendipine Triglyceride, Hot homogenization- 102123 Improved bioavailability (Manjunath and
Phosphatidylcholine 95%, ultrasonication method Venkateswarlu, 2006)
Poloxamer 188
SLN Nitrendipine Tripalmitin, Glyceryl Hot homogenization 110.6115.4 nm, 116.4122.3 nm, Improvement of oral bioavailability and reduction of rst-pass (Kumar et al., 2007)
Monostearate, Cetyl Palmitate ultrasonication 132.6137.4 nm metabolism
SLN Octa decylamine- Stearic acid, Polyethylene Solvent diusion method 202.7 4025 Improved bioavailability (30%) (Yuan et al., 2007a)
uorescein glycol monostearate
isothiocyanae
NLC Oridonon GMS/Medium chain Low temperature solidi 232.1 nm Prolonged drug release (Jia et al., 2010)
triglyceride(MCT) cation
SLN Otcadecylamine Stearic acid, Poloxamer 188, Solvent diusion 202.7 4.25 nm Enhancement of GI absorption, ymphatic transport and prolonged (Yuan et al., 2007a)
Otcadecylamine, release
Polyethylene glycol
monostearate
NLC Oxybenzone GMS, Miglyol 812, Oleic acid, Solvent diusion 327 30 to 797 100 Enhanced the sunscreen ecacy to about six fold while reducing its (Sanad et al., 2010)
PVA, Carbopol934P side eects.
NLC Oxybenzone GMS/Miglyol Solvent diusion method 0.5280.641 increased the in vitro sun protection factor and erythemal UVA (Sanad et al., 2010)
protection factor of oxybenzone more than 6- and 8-folds,
respectively, with fewer side eects
SLN Pentoxifylline Pluronic F-68, Soy Lecithin, Homogenization followed by 255 nm and 4 m Improvement of oral bioavailability and reduction of rst-pass (Varshosaz et al.,
Stearic Acid, Cetyl Alcohol, the ultrasonication metabolism 2010a)

45
Tween 20,
SLN Praziquantel Hydrogenated castor oil, Poly Hot homogenization and 344.0 15.1 nm, Improvement of bioavailability and prolonged systemic circulation (Xie et al., 2010)
vinyl alcohol, ultrasonication
NLC Progesterone Monostearin, Stearic Acid, Melt-emulsication technique 321.7485.5 nm Potential drug delivery system for oral administration (Yuan et al., 2007b)
Oleic Acid
NLC Psoralens Precirol/Squlaene High-pressure 300 and 200 nm Enhanced permeation and controlled release of the drug (Fang et al., 2008)
homogenization
SLN Puerarin Monostearin Soya lecithin, Solvent injection method 160 Improved bioavailability (Luo et al., 2011)
Poloxamer188
SLN Quercetin Glyceryl monostearate, Soya Emulsication-solidication 155.3 22.1 Improved bioavailability (Li et al., 2009)
lecithin, Tween-80 and PEG
400
NLC Quercetin GMS/MCT Emulsion evaporation solidi 215.2 nm Increased drug retention in epidermis and enhanced the therapeutic (Chen-Yu et al., 2012)
cation eect.
SLN Repaglinide Glycerol Monostearate, Modied solvent injection 175350 nm Well tolerated toxicity level (Rawat et al., 2011)
Tristearin method
NLC Resveratrol Compritol 888 ATO/Miglyol High shear homogenization 287.2 nm 5.1 and 110.5 nm 1.3, The drug loaded NLCs with smaller particle size and high drug (Gokce et al., 2012)
loading showed greater antioxidant activity as compared to SLNs.
SLN Rifampicin, Isoniazid Stearic Acid, Polyvinyl Emulsion-solvent diusion 70100 Improved bioavailability, stability and reducing dosing frequency (Pandey et al., 2005)
and Pyrazinamide Alcohol
NLC Tacrolimus GMS/Oleic Acid Hot sonication and 123.4 0.3 nm Penetration rate is 1.64 times increased than the commercial (Nam et al., 2011)
homogenization method tacrolimus ointment, Protopic
SLN Tobramycin Stearic acid Epikuron 200, Microemulsion 116 Improved bioavailability, sustained drug release and lymphatic (Cavalli et al., 2003)
Sodium taurocholate targeting
NLC Topotecan Stearic Acid/Oleic Acid Micro-emulsication 108168 nm Sustained release, improved chemical stability and cytotoxicity. (Souza et al., 2011)
technique
(continued on next page)
Sustainable Chemistry and Pharmacy 6 (2017) 3756
P. Ganesan, D. Narayanasamy Sustainable Chemistry and Pharmacy 6 (2017) 3756

temperature-induced drug degradation, b) drug distribution into the

(Arajo et al., 2011)


aqueous phase during homogenization, c) complexity of the crystal-

(Mei et al., 2005)

(Luo et al., 2006)


lization step of the nanoemulsion leading to several modications and/
or supercooled melts (Mehnert and Mder, 2001).
In Cold HPH, similar to hot homogenization, the solid lipid is he-
Ref.

ated, and drug molecules are incorporated into the matrix by dissolving
or dispersing them in molten lipid (Fig. 3b). The drug-containing lipid
Increase bioavailability, controlled release and decrease toxicity with

melt is rapidly solidied by cooling with dry ice or liquid nitrogen.


Improved oral BA by increased saturated solubility and reduced

Rapid cooling favours homogenous distribution of the drug within the


lipid material. The solid is then ground into a ne powder by milling
into microparticles. The microparticles are subsequently dispersed in a
cold aqueous surfactant solution. The dispersion is subjected to high-
pressure homogenization to generate SLNs. Cold homogenization in-
volves homogenization of solid lipids as opposed to a lipid melt in hot
homogenization. This dispersion of solid lipids requires high energy
input which in turn requires harsh homogenization conditions. Thus
homogenization itself is more eective for the hot case, and smaller
particles which are the more monodisperse result (Mder et al., 2004).

4.1.4. High shear homogenization and/or ultrasonication technique


High shear homogenization and ultrasonication are dispersing
Improved stability

protective eect

techniques (Fig. 4). Lipid nanoparticle dispersions are obtained by


dispersing the melted lipid in the warm aqueous phase containing
metabolism

surfactants by high sheer homogenization followed by ultrasonication.


This method primarily involves heating of a solid lipid to approximately
510 C above its melting point. The lipid melt is dispersed in an aqu-
eous surfactant solution at the same temperature under high speed
stirring to form an emulsion. Subsequent sonication reduces the droplet
size of the emulsion. Gradual cooling of the warm emulsion below the
crystallization temperature of the lipid yields a lipid nanoparticle dis-
persion. Concentrated lipid nanoparticle dispersions can be obtained by
ultracentrifugation (Shah et al., 2015).
Mechanism of particle formation

Shear between adjacent particles Formation, growth and implosive


173.30 0.32

collapse of bubbles due to cavitation forces.


70200 nm
Size (nm)

116.1 nm

4.2. Low energy approaches


High pressure homogenization

4.2.1. Microemulsion technique


Microemulsion formation is used as a stage in the production of SLN
and NLC since the early 90s (Gasco, 1993). In this method, the micro-
emulsion is spontaneously formed due to the high surfactants/lipid
Ultrasonic-solvent
Probe soniation

ratio (Fig. 5). The proportions of the excipients are essential, and in
emulsication

most cases, pseudo-ternary diagrams are used to study and describe the
Method

areas of microemulsion formation. This method is simple and is per-


formed by several common steps. Initially, the lipids are melted and
mixed with hot surfactant solution. Gentle stirring is applied until the
microemulsion is formed. In the second stage, the hot microemulsion is
Glyceryl monostearate, Soya
Poloxamine 908, Soybean

dispersed in a high volume of cold water (23 C) under moderate


Precirol ATO 5/Squalene

hydrogenated castor oil

stirring. This causes the liquid droplets to solidify. SLN or NLC obtained
Tristearin Glyceride,

lecithin, Tween80,

by this technique are spherical in shape and have narrow size dis-
Polyoxyethylene

tribution. However, the method suers from several drawbacks the


nal dispersion is very diluted (ranging between 1:25 up to 1:50 with
Excipients

Lecithin

respect to the hot emulsion). This may require further concentration by


ultraltration, lyophylization or other methods. The high concentration
of surfactants/co-surfactants used is another major disadvantage of this
technique (Gasco, 1993; Svilenov and Tzachev, 2014).
Mechanism of particle formation
Triamcinolone

Vinpocetine

Lipid crystallization due to rapid solidication of microemulsion.


Triptolide
acetonide
Table 4 (continued)

Drug

4.2.2. Membrane contactor technique


This method employs a cylindrical membrane module: aqueous
Type

NLC

SLN

SLN

phase containing a surfactant is circulated in internal channel of the


membrane, and melted lipid is pressed through pores of the membrane

46
P. Ganesan, D. Narayanasamy Sustainable Chemistry and Pharmacy 6 (2017) 3756

Fig. 3. Homogenization technique. a). Hot homogenization technique b). Cold homogenization technique.

into internal water ow allowing the formation of small droplets which (Shah et al., 2015). This technique mainly depends on the change in the
are swept away by the aqueous phase; water is maintained at lipid properties of polyoxyethylated surfactants at dierent temperatures
melting temperature. SLNs/NLCs are then formed by cooling the pre- (Fig. 7). The hydrophilic-lipophilic balance (HLB) value of surfactants
paration to room temperature (Charcosset et al., 2005) (Fig. 6). The dened by Grin is valid at 25 C. At this temperature, the hydrophilic
method is scalable, and the particle size can be tuned by using mem- parts of the surface-active compounds are hydrated to a certain extent.
branes with a dierent pore size (Svilenov and Tzachev, 2014). Moreover, the dehydration of the ethoxy groups occurs when an in-
Mechanism of particle formation crease in temperature. Thus, the lipophilicity of the molecules of the
surface-active compounds rises with the decrease in HLB value. At a
Lipid/oil phase infuses through membrane pores into the tangen- certain point, the surface-active compounds of the anity to the aqu-
tially owing aqueous phase to form droplets. eous and lipid phase is equal - this temperature is called as the phase
Oil droplets crystallize to form lipid nanoparticles. inversion temperature. This particulate state is characterized by very
low surface tension and the presence of complex structures in the
system. If the temperature is further increased the anity of the sur-
4.2.3. Phase inversion temperature (PIT) technique face-active compounds becomes higher enough to the lipid phase to
Transformation of an o/w type to a w/o type of emulsion is termed stabilize emulsions of W/O type.
phase inversion, can be induced by changing the temperature, and If cooled down the system goes through the reverse process. Due to
the temperature at which the inversion occurs is referred to as the PIT

Fig. 4. High shear homogenization/ultrasonication technique.

47
P. Ganesan, D. Narayanasamy Sustainable Chemistry and Pharmacy 6 (2017) 3756

Fig. 5. Micro emulsion technique.

with cold water causing phase inversion of the emulsion and breaking
resulting in SLNs/NLCs
Mechanism of particle formation

Spontaneous inversion of o/w emulsion to w/o emulsion due to


thermal treatment (subsequent heating- cooling cycles).
Lipid crystallization as a result of emulsion breakage due to irre-
versible shock induced by rapid cooling.

4.2.4. Coacervation technique


Lipid nanoparticles are produced by acidication of a micellar so-
lution of fatty acid alkaline salts (Battaglia et al., 2010; Bianco et al.,
2010; Chirio et al., 2011; Gallarate et al., 2010) (Fig. 8). Before pre-
paration of lipid nanoparticles, a stock solution of the polymeric sta-
bilizer is prepared by heating in hot water. A sodium salt of the fatty
acid is homogenously dispersed in the polymeric stabilizer stock solu-
tion, and the solution is heated above the Krat point of the sodium salt
of the fatty acid (Battaglia et al., 2014), under constant stirring, to
obtain a clear solution. The drug (solubilized in ethanol) is later
added to the clear solution, with constant stirring, until a single phase is
Fig. 6. Membrane contactor technique. obtained. The gradual addition of a coacervating solution (or on acid-
ifying the solution) to this mixture yields a suspension. Further cooling
the specic properties of the system around the PIT, very small particles of the suspension in a water bath, under constant agitation, yields drug-
are spontaneously formed just below that temperature. If rapid cooling loaded nanoparticles which are well dispersed (Shah et al., 2015;
is applied at this point stable particles with a desirable size and poly- Battaglia et al., 2010,).
dispersity can be obtained (Izquierdo et al., 2002). In this method, lipid, Mechanism of particle formation
drug, water and surfactant are mixed together on magnetic stirring,
three heating and cooling cycles are performed which is then diluted Decrease in pH of micellar solution of alkaline salts of fatty acids by
acidication (coacervating solution) in the presence of a polymeric

Fig. 7. Phase inversion temperature (PIT) technique.

48
P. Ganesan, D. Narayanasamy Sustainable Chemistry and Pharmacy 6 (2017) 3756

Fig. 8. Coacervation technique.

stabilizer causes proton exchange a lipid precipitation (coacerva- matrix is produced. This o/w micro emulsion is further cooled at room
tion). temperature or at 4 C so as to precipitate SLN from it. This method is
reproducible, simple and easy to scale up. Moreover, all the ingredients
used are biocompatible; no organic solvents are used in the preparation
4.2.5. Double emulsion technique
method.
The double emulsion technique in the preparation of SLN and NLC is
suitable for hydrophilic active pharmaceutical ingredients and peptides
(Svilenov and Tzachev, 2014). In this method, an aqueous solution of
4.3. Approaches with organic solvents
the drug is emulsied in melted lipid blend to form primary W/O
emulsion stabilized with suitable excipients (Fig. 9). The primary W/O
4.3.1. Emulsication-solvent evaporation technique (Pedersen et al., 2006)
emulsion is dispersed in aqueous solution of hydrophilic emulsier to
This technique involves three steps of preparation. Such as (i) pre-
form a double W/O/W emulsion. Then the double emulsion is stirred
paration of organic phase: lipophilic material is rst dissolved in an
and isolated by ltration. Relatively large particles are obtained with
appropriate volume of organic solvent by magnetic stirring. (ii) Pre-
this technique, but beside the incorporation of hydrophilic molecules, it
emulsication step: lipid-containing organic phase is dispersed in an
oers the possibility of surface modication, e.g. with PEGs.
appropriate volume of an aqueous solution using a high-speed homo-
Mechanism of particle formation
genizer in order to form a coarse pre-emulsion. (iii) Nano emulsication
step: the resulting coarse pre-emulsion is immediately passed through a
Lipid crystallization due to solidication of emulsion. high-pressure homogenizer at an operating pressure to obtain nano-
dispersion. Obtained nanodispersion is then kept on the magnetic
4.2.6. Micro emulsion cooling technique stirrer overnight, sometimes in a fume hood to drive o the organic
Recently, Mumper and Jay (Koziara et al., 2004, 2005; Mumper solvent. Upon solvent evaporation, nanodispersion is formed by pre-
et al., 2006) patented a micro emulsion based method for preparation cipitation of lipid material in the aqueous medium. Solidied nano-
of SLN. This method involves preparation of o/w micro emulsion where dispersion is then ltered through a sintered glass lter to remove lipid
an emulsifying wax is melted at 3755 C and addition of water which and drug agglomerates (Fig. 10). Nanoparticles obtained by this method
heated at the same temperature with minimal stirring so as to form a are small, monodisperse with high encapsulation eciency. The pro-
homogenous milky slurry. Further, after addition of specied amounts cess can be automated and scaled-up for the production of a large
of a suitable pharmaceutically acceptable polymeric surfactant in amount of nanoparticles (Jaiswal et al., 2004).
water, a stable and clear o/w micro emulsion in the form of a liquid Mechanism of particle formation

Fig. 9. Double emulsion technique.

49
P. Ganesan, D. Narayanasamy Sustainable Chemistry and Pharmacy 6 (2017) 3756

Fig. 10. Emulsication-solvent evaporation technique.

Lipid crystallization due to solvent evaporation in an anti-solvent.


4.3.2. Emulsication solvent diusion technique
Partially water-miscible solvents are used to solubilize the solid li-
pids. A number of solvents partially soluble in water such as benzyl
alcohol, butyl lactate, isobutyric acid, isovaleric acid and tetra-
hydrofuran have been used in the preparation of lipid nanoparticles
(Battaglia et al., 2007; Shahgaldian et al., 2003a, 2003b, 2003c). To
ensure initial thermodynamic equilibrium, organic solvents are satu-
rated with water. The transient oil- in-water emulsion is passed into
water under continuous stirring, which leads to solidication of dis-
persed phase forming lipid nanoparticles due to diusion of the organic
solvent. Typical emulsion: water ratios are 1:5 or 1:10 (Shah et al.,
2015) (Fig. 11).
Mechanism of particle formation:

Lipid crystallization due to diusion of solvent from internal organic


phase to external aqueous phase.
Fig. 12. Solvent injection technique.

4.3.3. Solvent injection technique


The basic principle of the solvent injection method is similar to the 1. Gradual solvent diusion out of lipid-solvent droplets into water
solvent diusion method. In the case of solvent injection method, lipids causes reduction of droplet size and simultaneously increases lipid
are dissolved in a water-miscible solvent (e.g. acetone, isopropanol, and concentration
methanol) or water-miscible solvent mixture and quickly injected into 2. Diusion of pure solvent from the lipid-solvent droplet causes local
an aqueous solution of surfactants through an injection needle variations in the interfacial tension at droplet surface, inducing re-
(Schubert and Mller-Goymann, 2003) (Fig. 12). Two simultaneous duction of size of droplets In this process particle size of SLNs/NLCs
eects contribute to the eective formation of SLNs/NLCs: can be inuenced and controlled by variation of process parameters

Fig. 11. Emulsication solvent diusion technique.

50
P. Ganesan, D. Narayanasamy Sustainable Chemistry and Pharmacy 6 (2017) 3756

such as injected solvent, lipid concentration, injected volume of consists in solubilizing CO2 in melted or liquid suspended substance(s)
solvent, lipid concentration in the solvent phase and viscosity of the (Reverchon and Porta, 2001), leading to a so-called gas-saturated so-
aqueous phase (Schubert and Mller-Goymann, 2003). lution/suspension that is further expanded through a nozzle with the
formation of solid particles or droplets. The advantage of this process is
Mechanism of particle formation: that the substances need not be soluble in CO2 (Byrappa et al., 2008).
In literature some examples of PGSS applied to lipid nano-and micro
Lipid crystallization due to rapid diusion of solvent from internal particles can be recovered. The most important are the so called Gas
organic phase to external aqueous phase. Assisted Melting Atomisation (GAMA) (Salmaso et al., 2009, Vezz
et al., 2010). Lipids are placed in a thermo stated mixing chamber (CM),
4.3.4. Supercritical uid (SCF) technique where they are melted and kept in contact with supercritical CO2 at
The process of preparing lipid nanoparticles from emulsions using selected temperature and pressure conditions. Then, the saturated lipid
SCF technology is referred to as supercritical uid extraction of uid is forced through the nozzle by opening the valve at the bottom of
emulsions (SFEE) (Chattopadhyay et al., 2006, 2007). The organic the CM, to produce micro particles. A CO2 reservoir is allowed to keep
solution is prepared by solubilizing the lipid material and the drug in an constant the pressure in the CM, and the ow rate through the nozzle as
organic solvent such as chloroform with the addition of a suitable well. Particles are gathered by a collection system and dispersed in
surfactant. The organic solution is dispersed into an aqueous solution water by vortexing and sonicating, to obtain suspensions. Polyethylene
(which may contain a co-surfactant), and the mixture is subsequently glycol (PEG) can be added to the formulation to increase the rate of
passed through a high-pressure homogenizer to form an o/w emulsion. dispersion in water.
The o/w emulsion is introduced from one end of the extraction column
(usually the top) at a constant ow rate, and the supercritical uid 5. Lyophilization
(maintained at constant temperature and pressure) is introduced
counter-currently at a constant ow rate. Lipid nanoparticle dispersions Stability of SLNs/NLCs mainly depends on the physicochemical
are formulated by continuous extraction of solvent from the o/w stability of solid lipid in their nano particles form, since they are more
emulsions (Shah et al., 2015, Svilenov and Tzachev, 2014) (Fig. 13). prone to degradation and coalescence. The physical stability of the SLN
Mechanism of particle formation: during prolonged storage can be determined by monitoring changes in
particle size, zeta potential, drug content, appearance, and viscosity as a
Parallel processes of supercritical uid extraction (diusion) of or- function of time (Wu et al., 2011; Schwarz et al., 1994; Mishra et al.,
ganic solvent from emulsions and lipid dissolution. 2012). In the case of chemical stability, the SLN ingredients should have
Expansion of organic phase; leads to lipid crystallization. a very narrow size distribution to avoid crystal growth by Ostwald ri-
pening (Mehnert and Mder, 2001). External parameters such as tem-
4.3.5. Particle from Gas Saturated Solution (PGSS) technique and Gas perature and light appear to be of primary importance for long-term
Assisted Melting Atomisation (GAMA) technique stability during the storage and shipping. In most cases, an increase in
PGSS involves melting of the material to be processed, which then particle size will be observed in a shorter period of time (Mishra et al.,
dissolves the SCF under pressure. The saturated solution is then ex- 2012).
panded across a nozzle where the SCF, which is more volatile, escapes, The zeta potential should, in general, remain higher than 60 mV
leaving dry ne particles. As the solubilities of compressed gases in li- for a dispersion to remain physically stable. This fact, in turn, is
quids and solids are usually high, and much higher than the solubilities achieved by lyophilization. Lyophilization is a promising way to in-
of such liquids and solids in the compressed gas phase, the process crease physical and chemical SLN stability over extended periods of

Fig. 13. Supercritical uid (SCF) technique.

51
P. Ganesan, D. Narayanasamy Sustainable Chemistry and Pharmacy 6 (2017) 3756

Table 5 cryoprotectants during the freezing step. Sugars isolate individual


Characterization of SLNs/ NLCs. particles in unfrozen fraction and thereby prevent particle aggregation
during freezing above Tg. Sugar vitrication is not required for this
Characterization parameters Analytical methods/
instrumentations eect (Allison et al., 2000; Pardeshi et al., 2012).
Similarly, water replacement hypothesis is a major mechanism for
Shape and surface morphology Transmission electron microscopy nanoparticle stabilization by lyoprotectants during the drying step.
(TEM)
These lyoprotectants are known to form hydrogen bonds with the sur-
Scanning electron microscopy (SEM)
Phase contrast optical microscopy face polar groups of nanoparticles and serve as water substitutes to
(PCM) preserve the native structure of nanoparticles, thereby stabilizing the
Atomic force microscopy (AFM) system (Pardeshi et al., 2012; Crowe et al., 1998).
Freeze fracture microscopy Freeze dried nanoparticles should have the following desirable
Vesicle size and size distribution Electron microscopy (SEM/TEM) characteristics:
Optical microscopy
Photon correlation spectroscopy (PCS)
(i) Preservation of primary physical and chemical characteristics of
Electrical surface potential and surface Zeta potential measurement the product.
pH pH-sensitive probes
(ii) Long-term stability.
Surface charge and electrophoretic Laser light scattering technique (iii) Acceptable relative humidity.
mobility

Surface hydrophobicity Hydrophobic interaction The efciency of the cryoprotectors decreases in the following
chromatography order: trehalose > sucrose > > glucose and maltose. The addition of
Two-phase partition
the time of the cryoprotector inuences the quality of the nal for-
Radiolabel probe
Contact angle measurement mulation. The cryoprotector was added to the sample when prior to
X-ray photoelectron spectroscopy homogenization the best results were obtained. The average particle
Synchrotron radiation X-ray (SAX) size remained almost unchanged under these circumstances (Swathi
Density Gas pycnometre et al., 2012). Storage over one year caused signicant increases in
Molecular weight Gel permeation chromatography (GPC) particle size (Mehnert and Mder, 2001). Besides, Schwarz et al. studied
Rheology Viscometer the eect of cryoprotectors during the lyophilization of SLN by using
In vitro release Dialysis membrane dissolution test
apparatus
glucose, mannose, maltose and trehalose in concentrations between
10% and 15% and reported the best results (Schwarz and Mehnert,
1997; Madden et al., 1985; Crowe et al., 1985; Mehnert and Mder,
2001). Cavalli et al. also reported in the case of trehalose that the 2%
time. Also, transformation into a solid form will prevent Ostwald ri- concentration of trehalose was insufcient to prevent lyophilization-
pening and avoid hydrolysis reactions (Mishra et al., 2012; Mehnert and induced particle aggregation and 15% concentration of trehalose was
Mder, 2001; Swathi et al., 2012). Stresses that destabilize the colloidal sufcient to produce particle sizes of around 100 nm and in poly-
nanoparticulate suspension may be generated during freeze drying, dispersity indices of 0.25 after reconstitution (Cavalli et al., 1997;
particularly stresses of freezing and dehydration. Aggregation and Mehnert and Mder, 2001).
sometimes an irreversible fusion of nanoparticles may destabilize the To transform an aqueous SLN dispersion into a dry product the
colloidal nanoparticulate system (Pardeshi et al., 2012). Obviously, two spray drying might be an alternative procedure to lyophilization. This
additional transformations between the formulations are necessary method has been used scarcely for SLN formulation (Hanumanaik et al.,
which might be the source of additional stability problems. The rst 2013), although spray drying is cheaper compared to lyophilization
transformation is from an aqueous dispersion to powder, which in- (Mehnert and Mder, 2001).
volves the freezing of the sample and the evaporation of water under
vacuum. Freezing of the sample might cause stability problems due to 6. Characterization of SLNs/ NLCs (Seetapan et al., 2010; Mller
the freezing out eect which results in changes of the osmolarity and et al., 2008; Silva et al., 2011, 2007; Subedi et al., 2009; Mishra
the pH. The second transformation is resolubilization, which involves, et al., 2012)
at least in its initial stages, situations which favor particle aggregation
(low water and high particle content, high osmotic pressure) (Mehnert Dierent techniques used for the characterization of SLNs/NLCs are
and Mder, 2001). tabulated in the Table 5.
To get rid of this problem, special excipients must be added before
freezing to protect the nanoparticulate suspension. The added ex- 7. Drug release
cipients that protect the system from freezing stress are called cryo-
protectants and those protecting from the drying stress are called lyo- Investigations of drug incorporation and release served as an im-
protectants (Pardeshi et al., 2012). The addition of cryoprotectors will portant tool in the design, development, and evaluation of potential
be necessary to decrease SLN aggregation and to obtain a better re- drug carrier systems (Pardeshi et al., 2012). Drugs incorporated into
dispersion of the dry product (Mehnert and Mder, 2001; Crowe et al., SLN are released by degradation and surface erosion of the lipid matrix
1986; Madden et al., 1985; Strauss et al., 1986; Hauser and Strauss, and by diusion of drug molecules through the lipid matrix (Mehnert
1988; Shulkin et al., 1984). Typical cryoprotective agents are sorbitol, and Mder, 2001). Drug release from SLN is mainly aected by loca-
mannose, trehalose, glucose, and polyvinylpyrroli- done. They decrease lization of the drug (Muchow et al., 2008):
the osmotic activity of water and crystallization and favor the glassy
state of the frozen sample (Shiau, 1990; Phan and Tso, 2001; Porter and 1. Localization of the drug within the core of solid lipid matrix oers
Charman, 2001; O'driscoll and Grin, 2008; Porter et al., 2007). the possibility to obtain a prolonged drug release.
Cryoprotectors are place holders which prevent the contact between 2. Localization of drug molecules on particle surface often leads to
discrete lipid nanoparticles. Furthermore, they inter-act with the polar burst eect (fast initial drug release). SLN can show a biphasic drug
head groups of the surfactants and serve as a kind of pseudo hydration release prole: an initial burst release, due to the drug localized at
shell (Mobley and Schreier, 1994). In addition, particle isolation hy- the surface, is followed by a more gradual release due to the drug
pothesis is one of the mechanisms for nanoparticle stabilization by localized in the lipid matrix.

52
P. Ganesan, D. Narayanasamy Sustainable Chemistry and Pharmacy 6 (2017) 3756

Therefore, the extent of burst release can be controlled by control- using the high-pressure homogenization method with existing ma-
ling drug solubility in the aqueous phase during production, which, in chinery, ease of large-scale manufacturing and avoidance of toxicity
turn, can be controlled via the temperature employed and the surfac- makes the lipid nanoparticle as a suitable condition for drug de-
tant concentration used. Higher temperature and higher surfactant livery and potentially attractive and marketable choices.
concentration increase the burst whereas production at room tem-
perature avoids partitioning of the drug into a water phase and sub- 9. Conclusion
sequent re-partitioning into lipid phase, thereby showing no burst re-
lease at all. To avoid or minimize the burst release, SLN can be From the review, it was evident that the various research groups are
produced surfactant-free or with surfactants unable to solubilize the increasingly attracted with lipid nanoparticles (SLNs & NLCs) due to its
drug (Muller et al., 2000; Pardeshi et al., 2012). The possible presence unique properties and a lot of advantages over traditional dosage forms
of alternative colloidal species always has to be taken into account for and their colloidal counterpoints. Hence, lipid nanoparticles are pro-
drug release characterization: stabilizing agent cannot be localized mising drug delivery system for the formulation of poorly water soluble
exclusively on the lipid surface as expected, but also in the aqueous drug in the pharmaceutical industry.
phase forming micelles, mixed micelles or liposomes that can solubilize
the drugs and constitute alternative drug incorporation sites (Mehnert Conicts of interest
and Mder, 2001).
The authors declare no conict of interest.
8. Links to green and sustainable chemistry
References
Nowadays the poorly water-soluble drug candidates are challenging
candidates not only in the formulation aspect but also for conducting Abdelbary, G., Fahmy, R.H., 2009. Diazepam-loaded solid lipid nanoparticles: design and
pharmacological, toxicological and pharmacokinetic studies during the characterization. Aaps Pharmscitech 10, 211219.
Agrawal, Y., Petkar, K.C., Sawant, K.K., 2010. Development, evaluation and clinical
drug development stage and in biological conditions. Also, it is mainly studies of acitretin loaded nanostructured lipid carriers for topical treatment of
responsible for toxicity problem associated with the drug design, de- psoriasis. Int. J. Pharm. 401, 93102.
velopment, optimization and in the biological fate of the drug in the Ahmed El-Harati, A., Charcosset, C., Fessi, H., 2006. Inuence of the formulation for solid
lipid nanoparticles prepared with a membrane contactor. Pharm. Dev. Technol. 11,
body. But all these problems can be overcome by formulating them as a 153157.
lipid nanoparticles which facilitates the absorption into the systemic Alex, M.A., Chacko, A., Jose, S., Souto, E., 2011. Lopinavir loaded solid lipid nano-
circulation through the intestinal lymphatic pathway and avoiding the particles (SLN) for intestinal lymphatic targeting. Eur. J. Pharm. Sci. 42, 1118.
All Rights Are Reserved by Mrs., Gupta, D.R., 2009. Solubility Enhancement by Solid
rst-pass eect, also facilitates the below-mentioned advantages which Lipid Nanoparticle.
is related to green chemistry. Allison, S.D., Dc Molina, M., Anchordoquy, T.J., 2000. Stabilization of lipid/DNA com-
plexes during the freezing step of the lyophilization process: the particle isolation

When compared to synthetic, semisynthetic excipients used in the hypothesis. Biochim. Biophys. Acta (BBA)-Biomembr. 1468, 127138.
Almeida, A.J., Runge, S., Mller, R.H., 1997. Peptide-loaded solid lipid nanoparticles
formulation of pharmaceutical formulations, the excipients used in (SLN): inuence of production parameters. Int. J. Pharm. 149, 255265.
this lipid nanoparticles are composed of physiological or physiolo- Anastas, P.T., Warner, J.C., 1998. Green Chemistry Theory and Practice. Oxford
gically related lipids. Hence, the pathways for absorption, metabo- University Press, New York, pp. 30.
Angrick, M., Kmmerer, K., Meinzer, L., 2006. Nachhaltige Chemie: erfahrungen und
lism and transportation are present in the body, which may con- Perspektiven. Metropolis Verlag fr konomie, Ges. Polit.
tribute to a large extent to the bio fate of lipidic carrier. Anton, N., Benoit, J.-P., Saulnier, P., 2008. Design and production of nanoparticles for-

Furthermore, the excipients of lipid nanoparticles (lipid, surfactants mulated from nano-emulsion templatesa review. J. Control. Release 128, 185199.
Arajo, J., Nikolic, S., Egea, M.A., Souto, E.B., Garcia, M.L., 2011. Nanostructured lipid
and cosolvents) have important role in physiology of the body, such carriers for triamcinolone acetonide delivery to the posterior segment of the eye.
as energy storage (lipids), a part of biomembranes (phospholipids), Colloids Surf. B: Biointerfaces 88, 150157.
and have key function in metabolism (bile acids), hence considered Basavaraj, K., 2012. Nanotechnology in medicine and relevance to dermatology: present
concepts. Indian J. Dermatol. 57, 169.
as harmless and usually scheduled in the generally recognized as Battaglia, L., Gallarate, M., 2012. Lipid nanoparticles: state of the art, new preparation
safe category which is US FDA-approved. methods and challenges in drug delivery. Expert Opin. Drug Deliv. 9, 497508.
Since the lipid nanoparticles composed of physiological lipids and Battaglia, L., Trotta, M., Gallarate, M., Carlotti, M.E., Zara, G.P., Bargoni, A., 2007. Solid
lipid nanoparticles formed by solvent-in-water emulsiondiusion technique: devel-
its digestion and absorption is similar to physiological lipids, it not
opment and inuence on insulin stability. J. Microencapsul. 24, 672684.
possess any toxic eects and decreases the adverse side eects of the Battaglia, L., Gallarate, M., Cavalli, R., Trotta, M., 2010. Solid lipid nanoparticles pro-
drug delivery systems when compared to other of metal or poly- duced through a coacervation method. J. Microencapsul. 27 (1), 7885.
Battaglia, L., Gallarate, M., Panciani, P.P., Ugazio, E., Sapino, S., Peira, E., Chirio, D.,
meric nature. Due to its biodegradation and biocompatible nature, it
2014. Techniques for the preparation of solid lipid nano and microparticles. In: Ali
secures the title of "Nano safe carrier". Demir Sezer, (Ed.), Application of Nanotechnology in Drug Delivery. InTech.
Moreover, in the formulation point of view, lipid nanoparticles are Benita, S., Bhm, B.H., 1998. Emulsions and Nanosuspensions for the Formulation of
Poorly Soluble Drugs. CRC Press.
much easier to manufacture than bio polymeric nanoparticles. Also,
Bevilacqua, A., Cibelli, F., Corbo, M.R., Sinigaglia, M., 2007. Eects of highpressure
it possess many advantages such as homogenization on the survival of Alicyclobacillus acidoterrestris in a laboratory
medium. Lett. Appl. Microbiol. 45 (4), 382386.
Good production scalability (sustainability). Bianco, M., Gallarate, M., Trotta, M., Battaglia, L., 2010. Amphotericin B loaded SLN
prepared with the coacervation technique. J. Drug Deliv. Sci. Technol. 20, 187191.
Avoidance of organic solvents in the formulation process. Bunjes, H., Westesen, K., Koch, M.H., 1996. Crystallization tendency and polymorphic
No special solvent required. transitions in triglyceride nanoparticles. Int.J. Pharm. 129, 159173.
Conventional emulsion manufacturing methods applicable. Byrappa, K., Ohara, S., Adschiri, T., 2008. Nanoparticles synthesis using supercritical
uid technologytowards biomedical applications. Adv. Drug Deliv. Rev. 60 (3),
Easy to scale up and sterilize. 299327.
Industrial adaptability. Cannon, A.S., Warner, J.C., 2011. The science of green chemistry and its role in educa-
tional reform. New Solut. 21 (3), 499517.

Also, lipid nanoparticles combine the attributes of both nanosized Cavalli, R., Caputo, O., Carlotti, M.E., Trotta, M., Scarnecchia, C., Gasco, M.R., 1997.
Sterilization and freeze-drying of drug-free and drug-loaded solid lipid nanoparticles.
particles and lipid carriers to improve the bioavailability of active Int. J. Pharm. 148, 4754.
pharmaceutical ingredients (APIs). Cavalli, R., Bargoni, A., Podio, V., Muntoni, E., Zara, G.P., Gasco, M.R., 2003. Duodenal

From all these advantages, such as preparation by using physiolo- administration of solid lipid nanoparticles loaded with dierent percentages of to-
bramycin. J. Pharm. Sci. 92, 10851094.
gical or physiologically related lipids which simulate physiological Charcosset, C., El-Harati, A., Fessi, H., 2005. Preparation of solid lipid nanoparticles using
lipids, lack of organic solvent usage during the production processes a membrane contactor. J. Control. Release 108, 112120.

53
P. Ganesan, D. Narayanasamy Sustainable Chemistry and Pharmacy 6 (2017) 3756

Chattopadhyay, P., Hu, R., Shekunov, B.Y., 2006. Drug encapsulation using supercritical inversion-based process for the preparation of lipid nanocarriers. Pharm. Res. 19,
uid extraction of emulsions. J. Pharm. Sci. 95, 667679. 875880.
Chattopadhyay, P., Shekunov, B.Y., Yim, D., Cipolla, D., Boyd, B., Farr, S., 2007. Hu, F., Hong, Y., Yuan, H., 2004a. Preparation and characterization of solid lipid nano-
Production of solid lipid nanoparticle suspensions using supercritical uid extraction particles containing peptide. Int. J. Pharm. 273, 2935.
of emulsions (SFEE) for pulmonary delivery using the AERx system. Adv. Drug Deliv. Hu, L., Tang, X., Cui, F., 2004b. Solid lipid nanoparticles (SLNs) to improve oral bioa-
Rev. 59, 444453. vailability of poorly soluble drugs. J. Pharm. Pharmacol. 56, 15271535.
Chen, C.-C., Tsai, T.-H., Huang, Z.-R., Fang, J.-Y., 2010. Eects of lipophilic emulsiers on Hu, L., Xing, Q., Meng, J., Shang, C., 2010. Preparation and enhanced oral bioavailability
the oral administration of lovastatin from nanostructured lipid carriers: physico- of cryptotanshinone-loaded solid lipid nanoparticles. Aaps Pharmscitech 11,
chemical characterization and pharmacokinetics. Eur. J. Pharm. Biopharm. 74, 582587.
474482. Huang, Z.R., 2008. Development and evaluation of lipid nanoparticles for camptothecin
Chen, G., Hou, S., Hu, P., Hu, Q., Guo, D., Xiao, Y., 2008. In vitro dexamethasone release delivery: a comparison of solid lipid nanoparticles, nanostructured lipid carriers, and
from nanoparticles and its pharmacokinetics in the inner ear after administration of lipid emulsion. Acta Pharmacol. Sin. 29, 10941102.
the drug-loaded nanoparticles via the round window. Nan Fang yi ke da xue xue bao Iqbal, M.A., Md, S., Sahni, J.K., Baboota, S., Dang, S., Ali, J., 2012. Nanostructured lipid
= J. South. Med. Univ. 28, 10221024. carriers system: recent advances in drug delivery. J. Drug Target. 20, 813830.
Chen-Yu, G., Chun-Fen, Y., Qi-Lu, L., Qi, T., Yan-Wei, X., Wei-Na, L., Guang-Xi, Z., 2012. Izquierdo, P., Esquena, J., Tadros, T.F., Dederen, C., Garcia, M., Azemar, N., Solans, C.,
Development of a quercetin-loaded nanostructured lipid carrier formulation for to- 2002. Formation and stability of nano-emulsions prepared using the phase inversion
pical delivery. Int. J. Pharm. 430, 292298. temperature method. Langmuir 18, 2630.
Chirio, D., Gallarate, M., Peira, E., Battaglia, L., Serpe, L., Trotta, M., 2011. Formulation Jaiswal, J., Gupta, S.K., Kreuter, J., 2004. Preparation of biodegradable cyclosporine
of curcumin-loaded solid lipid nanoparticles produced by fatty acids coacervation nanoparticles by high-pressure emulsication-solvent evaporation process. J.
technique. J. Microencapsul. 28, 537548. Control. Release 96, 169178.
Cirri, M., Bragagni, M., Mennini, N., Mura, P., 2012. Development of a new delivery Jaiswal, P., Gidwani, B., Vyas, A., 2016. Nanostructured lipid carriers and their current
system consisting in drugin cyclodextrinin nanostructured lipid carriers for ke- application in targeted drug delivery. Artif. Cells Nanomed. Biotechnol. 44, 2740.
toprofen topical delivery. Eur. J. Pharm. Biopharm. 80, 4653. Jenning, V., Thnemann, A.F., Gohla, S.H., 2000. Characterisation of a novel solid lipid
Conlin, A.K., Seidman, A.D., Bach, A., Lake, D., Dickler, M., D'andrea, G., Traina, T., nanoparticle carrier system based on binary mixtures of liquid and solid lipids. Int. J.
Danso, M., Brufsky, A.M., Saleh, M., 2010. Phase II trial of weekly nanoparticle al- Pharm. 199, 167177.
bumin-bound paclitaxel with carboplatin and trastuzumab as rst-line therapy for Jensen, L.B., Magnussson, E., Gunnarsson, L., Vermehren, C., Nielsen, H.M., Petersson, K.,
women with HER2-overexpressing metastatic breast cancer. Clin. Breast Cancer 10, 2010. Corticosteroid solubility and lipid polarity control release from solid lipid
281287. nanoparticles. Int. J. Pharm. 390, 5360.
Crowe, J.H., Carpenter, J.F., Crowe, L.M., 1998. The role of vitrication in anhydrobiosis. Jia, L., Zhang, D., Li, Z., Duan, C., Wang, Y., Feng, F., Wang, F., Liu, Y., Zhang, Q., 2010.
Annu. Rev. Physiol. 60, 73103. Nanostructured lipid carriers for parenteral delivery of silybin: biodistribution and
Crowe, L.M., Crowe, J.H., Rudolph, A., Womersley, C., Appel, L., 1985. Preservation of pharmacokinetic studies. Colloids Surf. B: Biointerfaces 80, 213218.
freeze-dried liposomes by trehalose. Arch. Biochem. Biophys. 242, 240247. Jorgensen, L., Nielson, H.M., 2009. Delivery Technologies for Biopharmaceuticals:
Crowe, L.M., Womersley, C., Crowe, J.H., Reid, D., Appel, L., Rudolph, A., 1986. Peptides, Proteins, Nucleic Acids and Vaccines. John Wiley & Sons Ltd, Chichester,
Prevention of fusion and leakage in freeze-dried liposomes by carbohydrates. United Kingdom.
Biochim. Biophys. Acta (BBA)-Biomembr. 861, 131140. Kakkar, V., Singh, S., Singla, D., Kaur, I.P., 2011. Exploring solid lipid nanoparticles to
Das, S., Chaudhury, A., 2011. Recent advances in lipid nanoparticle formulations with enhance the oral bioavailability of curcumin. Mol. Nutr. Food Res. 55, 495503.
solid matrix for oral drug delivery. Aaps Pharmscitech 12, 6276. Kang, K.W., Chun, M.-K., Kim, O., Subedi, R.K., Ahn, S.-G., Yoon, J.-H., Choi, H.-K., 2010.
Doktorovov, S., Arajo, J., Garcia, M.L., Rakovsk, E., Souto, E.B., 2010. Formulating Doxorubicin-loaded solid lipid nanoparticles to overcome multidrug resistance in
uticasone propionate in novel PEG-containing nanostructured lipid carriers (PEG- cancer therapy. Nanomed.: Nanotechnol. Biol. Med. 6, 210213.
NLC). Colloids Surf. B: Biointerfaces 75, 538542. Kaur, Sarabjot, Nautyal, Ujjwal, Singh, Ramandeep, Singh, Satvinder, Devi, Anita, 2015.
Domingo, C., Saurina, J., 2012. An overview of the analytical characterization of na- Nanostructure lipid carrier (NLC): the new generation of lipid nanoparticles. Asian
nostructured drug delivery systems: towards green and sustainable pharmaceuticals: Pac. J. Health Sci. 2 (2), 7693.
a review. Anal. Chim. Acta 744, 822. Kayser, O., Lemke, A., Hernandez-Trejo, N., 2005. The impact of nanobiotechnology on
Emeje, M.O., Akpabio, E.I., Obidike, I.C., Ofoefule, S.I., 2012. Nanotechnology in Drug the development of new drug delivery systems. Curr. Pharm. Biotechnol. 6, 35.
Delivery. INTECH Open Access Publisher. Kheradmandnia, S., Vasheghani-Farahani, E., Nosrati, M., Atyabi, F., 2010. Preparation
Fang, J.-Y., Fang, C.-L., Liu, C.-H., Su, Y.-H., 2008. Lipid nanoparticles as vehicles for and characterization of ketoprofen-loaded solid lipid nanoparticles made from
topical psoralen delivery: solid lipid nanoparticles (SLN) versus nanostructured lipid beeswax and carnauba wax. Nanomed.: Nanotechnol. Biol. Med. 6, 753759.
carriers (NLC). Eur. J. Pharm. Biopharm. 70, 633640. Konwarh, R., Pramanik, S., Kalita, D., Mahanta, C.L., Karak, N., 2012. Ultrasonicationa
Freitas, C., Mller, R., 1999. Correlation between long-term stability of solid lipid na- complementary green chemistrytool to biocatalysis: a laboratory-scale study of ly-
noparticles (SLN) and crystallinity of the lipid phase. Eur. J. Pharm. Biopharm. 47, copene extraction. Ultrason. Sonochem. 19 (2), 292299.
125132. Koziara, J.M., Lockman, P.R., Allen, D.D., Mumper, R.J., 2004. Paclitaxel nanoparticles
Gaba, B., Fazil, M., Ali, A., Baboota, S., Sahni, J.K., Ali, J., 2015. Nanostructured lipid for the potential treatment of brain tumors. J. Control. Release 99 (2), 259269.
(NLCs) carriers as a bioavailability enhancement tool for oral administration. Drug Koziara, J.M., Oh, J.J., Akers, W.S., Ferraris, S.P., Mumper, R.J., 2005. Blood compat-
Del. 22, 691700. ibility of cetyl alcohol/polysorbate-based nanoparticles. Pharm. Res. 22 (11),
Galitsky, C., Chan, S.-C., Worrell, E., Masanet, E., 2008. Energy Eciency Improvement 18211828.
and Cost Saving Opportunities for the Pharmaceutical Industry. An ENERGY STAR Kumar, M., 2000. Nano and microparticles as controlled drug delivery devices. J. Pharm.
Guide for Energy and Plant Managers (Report LBNL-57260). Lawrence Berkeley Pharm. Sci. 3, 234258.
National Laboratory, Berkeley, California. Kumar, S., Dilbaghi, N., Saharan, R., Bhanjana, G., 2012. Nanotechnology as emerging
Gallarate, M., Trotta, M., Battaglia, L., Chirio, D., 2010. Cisplatin-loaded SLN produced by tool for enhancing solubility of poorly water-soluble drugs. BioNanoScience 2,
coacervation technique. J. Drug Deliv. Sci. Technol. 20, 343347. 227250.
Gasco, M.R., 1993. Method for producing solid lipid microspheres having a narrow size Kumar, V.V., Chandrasekar, D., Ramakrishna, S., Kishan, V., Rao, Y.M., Diwan, P.V.,
distribution. Google Patents. 2007. Development and evaluation of nitrendipine loaded solid lipid nanoparticles:
Gokce, E.H., Korkmaz, E., Dellera, E., Sandri, G., Bonferoni, M.C., Ozer, O., 2012. inuence of wax and glyceride lipids on plasma pharmacokinetics. Int. J. Pharm. 335,
Resveratrol-loaded solid lipid nanoparticles versus nanostructured lipid carriers: 167175.
evaluation of antioxidant potential for dermal applications. Int J. Nanomed. 7, Kmmerer, K., Hempel, M., 2010. Green and Sustainable Pharmacy. Springer, Berlin.
18411850. Kuo, Y.-C., Chung, J.-F., 2011. Physicochemical properties of nevirapine-loaded solid
Gonzalez-Mira, E., Egea, M., Garcia, M., Souto, E., 2010. Design ocular tolerance of lipid nanoparticles and nanostructured lipid carriers. Colloids Surf. B: Biointerfaces
urbiprofen loaded ultrasound-engineered NLC. Colloids Surf. B: Biointerfaces 81, 83, 299306.
412421. Lander, R., Manger, W., Scouloudis, M., Ku, A., Davis, C., Lee, A., 2000. Gaulin homo-
Gupta, M., Vyas, S.P., 2012. Development, characterization and in vivo assessment of genization: a mechanistic study. Biotechnol. Progress. 16, 8085.
eective lipidic nanoparticles for dermal delivery of uconazole against cutaneous Li, H., Zhao, X., Ma, Y., Zhai, G., Li, L., Lou, H., 2009. Enhancement of gastrointestinal
candidiasis. Chem. Phys. Lipids 165, 454461. absorption of quercetin by solid lipid nanoparticles. J. Control. Release 133,
Gursoy, R.N., Benita, S., 2004. Self-emulsifying drug delivery systems (SEDDS) for im- 238244.
proved oral delivery of lipophilic drugs. Biomed. Pharmacother. 58, 173182. Lim, S.-J., Kim, C.-K., 2002. Formulation parameters determining the physicochemical
Hanafy, A., Spahn-Langguth, H., Vergnault, G., Grenier, P., Grozdanis, M.T., Lenhardt, T., characteristics of solid lipid nanoparticles loaded with all-trans retinoic acid. Int. J.
Langguth, P., 2007. Pharmacokinetic evaluation of oral fenobrate nanosuspensions Pharm. 243, 135146.
and SLN in comparison to conventional suspensions of micronized drug. Adv. Drug Lin, Y.-K., Huang, Z.-R., Zhuo, R.-Z., Fang, J.-Y., 2010. Combination of calcipotriol and
Deliv. Rev. 59, 419426. methotrexate in nanostructured lipid carriers for topical delivery. Int J. Nanomed. 5,
Hanumanaik, M., Patel, S.K., Sree, K.R., 2013. Solid lipid nanoparticles; A review. IJPSR 4 117128.
(3), 928940. Lippacher, A., Mller, R., Mder, K., 2000. Investigation on the viscoelastic properties of
Harde, H., Das, M., Jain, S., 2011. Solid lipid nanoparticles: an oral bioavailability en- lipid based colloidal drug carriers. Int. J. Pharm. 196, 227230.
hancer vehicle. Expert Opin. Drug Deliv. 8, 14071424. Liu, C., Liu, D., Bai, F., Zhang, J., Zhang, N., 2010a. In vitro and in vivo studies of lipid-
Hauser, H., Strauss, G., 1988. Stabilization of Small, Unilamellar Phospholipid Vesicles by based nanocarriers for oral N3-o-toluyl-uorouracil delivery. Drug Deliv. 17,
Sucrose During Freezing and Dehydration. Biotechnological Applications of Lipid 352363.
Microstructures. Springer, Boston, MA, US, pp. 7180. Liu, C.-H., Wu, C.-T., 2010. Optimization of nanostructured lipid carriers for lutein de-
Heurtault, B., Saulnier, P., Pech, B., Proust, J.-E., Benoit, J.-P., 2002. A novel phase livery. Colloids Surf. A: Physicochem. Eng. Asp. 353, 149156.

54
P. Ganesan, D. Narayanasamy Sustainable Chemistry and Pharmacy 6 (2017) 3756

Liu, D., Liu, C., Zou, W., Zhang, N., 2010b. Enhanced gastrointestinal absorption of N3-O- low aqueous solubilitythe potential impact of lipid-based formulations. Adv. Drug
toluyl-uorouracil by cationic solid lipid nanoparticles. J. Nanopart. Res. 12, Deliv. Rev. 60, 617624.
975984. Paliwal, R., Rai, S., Vaidya, B., Khatri, K., Goyal, A.K., Mishra, N., Mehta, A., Vyas, S.P.,
Liu, D., Liu, Z., Wang, L., Zhang, C., Zhang, N., 2011. Nanostructured lipid carriers as 2009. Eect of lipid core material on characteristics of solid lipid nanoparticles de-
novel carrier for parenteral delivery of docetaxel. Colloids Surf. B: Biointerfaces 85, signed for oral lymphatic delivery. Nanomed.: Nanotechnol. Biol. Med. 5, 184191.
262269. Pandey, R., Sharma, S., Khuller, G., 2005. Oral solid lipid nanoparticle-based anti-
Luo, C.-F., Yuan, M., Chen, M.-S., Liu, S.-M., Zhu, L., Huang, B.-Y., Liu, X.-W., Xiong, W., tubercular chemotherapy. Tuberculosis 85, 415420.
2011. Pharmacokinetics, tissue distribution and relative bioavailability of puerarin Pardeike, J., Weber, S., Haber, T., Wagner, J., Zar, H., Plank, H., Zimmer, A., 2011.
solid lipid nanoparticles following oral administration. Int. J. Pharm. 410, 138144. Development of an itraconazole-loaded nanostructured lipid carrier (NLC) formula-
Luo, Y., Chen, D., Ren, L., Zhao, X., Qin, J., 2006. Solid lipid nanoparticles for enhancing tion for pulmonary application. Int. J. Pharm. 419, 329338.
vinpocetine's oral bioavailability. J. Control. Release 114, 5359. Pardeshi, C., Rajput, P., Belgamwar, V., Tekade, A., Patil, G., Chaudhary, K., Sonje, A.,
Ma, P., Dong, X., Swadley, C.L., Gupte, A., Leggas, M., Ledebur, H.C., Mumper, R.J., 2009. 2012. Solid lipid based nanocarriers: an overview/Nanonosai na bazi vrstih lipida:
Development of idarubicin and doxorubicin solid lipid nanoparticles to overcome pregled. Acta Pharm. 62, 433472.
Pgp-mediated multiple drug resistance in leukemia. J. Biomed. Nanotechnol. 5, Pathak, K., Raghuvanshi, S., 2015. Oral bioavailability: issues and solutions via nano-
151161. formulations. Clin. Pharmacokinet. 54, 325357.
Madden, T.D., Bally, M.B., Hope, M.J., Cullis, P.R., Schieren, H.P., Jano, A.S., 1985. Pathak, P., Nagarsenker, M., 2009a. Formulation and evaluation of lidocaine lipid na-
Protection of large unilamellar vesicles by trehalose during dehydration: retention of nosystems for dermal delivery. Aaps Pharmscitech 10, 985.
vesicle contents. Biochim. Biophys. Acta (BBA)-Biomembr. 817, 6774. Pathak, P., Nagarsenker, M., 2009b. Formulation and evaluation of lidocaine lipid na-
Mder, K., Mehnert, W., Nastruzzi, C., 2004. Solid Lipid Nanoparticlesconcepts, nosystems for dermal delivery. Aaps Pharmscitech 10, 985992.
Procedures, and Physicochemical Aspect. Lipospheres in Drug Targets and Delivery: Pedersen, N., Hansen, S., Heydenreich, A.V., Kristensen, H.G., Poulsen, H.S., 2006. Solid
Approaches, methods, and Applications. CRC Press, Boca Raton, pp. 122. lipid nanoparticles can eectively bind DNA, streptavidin and biotinylated ligands.
Malik, P., Shankar, R., Malik, V., Sharma, N., Mukherjee, T.K., 2014. Green chemistry Eur. J. Pharm. Biopharm. 62, 155162.
based benign routes for nanoparticle synthesis. J. Nanopart. Phan, C.T., Tso, P., 2001. Intestinal lipid absorption and transport. Front. Biosci.: J.
Manjunath, K., Venkateswarlu, V., 2005. Pharmacokinetics, tissue distribution and Virtual Libr. 6, D299D319.
bioavailability of clozapine solid lipid nanoparticles after intravenous and in- Poovi, G., 2016. Challenges and advances in solid lipid nanoparticle drug delivery sys-
traduodenal administration. J. Control. Release 107, 215228. tems. J. Pharm. Care Health Syst. 3 (Suppl.), S2.
Manjunath, K., Venkateswarlu, V., 2006. Pharmacokinetics, tissue distribution and Porter, C.J., Charman, W.N., 2001. Intestinal lymphatic drug transport: an update. Adv.
bioavailability of nitrendipine solid lipid nanoparticles after intravenous and in- Drug Deliv. Rev. 50, 6180.
traduodenal administration. J. Drug Target. 14, 632645. Porter, C.J., Trevaskis, N.L., Charman, W.N., 2007. Lipids and lipid-based formulations:
Manjunath, K., Reddy, J.S., Venkateswarlu, V., 2005. Solid lipid nanoparticles as drug optimizing the oral delivery of lipophilic drugs. Nat. Rev. Drug Discov. 6, 231248.
delivery systems. Methods Find. Exp. Clin. Pharmacol. 27, 127144. Potta, S.G., Minemi, S., Nukala, R.K., Peinado, C., Lamprou, D.A., Urquhart, A.,
Markarian, J., 2016. Reducing the environmental impact of manufacturing. Pharm. Douroumis, D., 2011. Preparation and characterization of ibuprofen solid lipid na-
Technol. 40, 3638. noparticles with enhanced solubility. J. Microencapsul. 28, 7481.
Martins, S., Sarmento, B., Ferreira, D.C., Souto, E.B., 2007. Lipid-based colloidal carriers Priano, L., Zara, G.P., El-Assawy, N., Cattaldo, S., Muntoni, E., Milano, E., Serpe, L.,
for peptide and protein delivery-liposomes versus lipid nanoparticles. Int. J. Musicanti, C., Prot, C., Gasco, M.R., 2011. Baclofen-loaded solid lipid nanoparticles:
Nanomed. 2, 595. preparation, electrophysiological assessment of ecacy, pharmacokinetic and tissue
Martins, S., Silva, A., Ferreira, D., Souto, E., 2009. Improving oral absorption of samon distribution in rats after intraperitoneal administration. Eur. J. Pharm. Biopharm. 79,
calcitonin by trimyristin lipid nanoparticles. J. Biomed. Nanotechnol. 5, 7683. 135141.
Medina-Gonzlez, Y., Aimar, P., Lahitte, J.F., Remigy, J.C., 2011. Towards green mem- Puglia, C., Sarpietro, M.G., Bonina, F., Castelli, F., Zammataro, M., Chiechio, S., 2011.
branes: preparation of cellulose acetate ultraltration membranes using methyl lac- Development, characterization, and in vitro and in vivo evaluation of benzocaineand
tate as a biosolvent. Int. J. Sustain. Eng. 4 (01), 7583. lidocaineloaded nanostructrured lipid carriers. J. Pharm. Sci. 100, 18921899.
Mehnert, W., Mder, K., 2001. Solid lipid nanoparticles: production, characterization and Puri, A., Loomis, K., Smith, B., Lee, J.-H., Yavlovich, A., Heldman, E., Blumenthal, R.,
applications. Adv. Drug Deliv. Rev. 47, 165196. 2009. Lipid-based nanoparticles as pharmaceutical drug carriers: from concepts to
Mei, Z., Li, X., Wu, Q., Hu, S., Yang, X., 2005. The research on the anti-inammatory clinic. Crit. Rev. Ther. Drug Carr. Syst. 26.
activity and hepatotoxicity of triptolide-loaded solid lipid nanoparticle. Pharmacol. Rajagopal, R., 2014. Sustainable Value Creation in the Fne and Speciality Chemicals
Res. 51, 345351. Industry. John Wiley & Sons Ltd., United kingdom.
Mishra, D.K., Dhote, V., Bhatnagar, P., Mishra, P.K., 2012. Engineering solid lipid na- Rao, K.K., 2008. Polymerized solid lipid nanoparticles for oral or mucosal delivery of
noparticles for improved drug delivery: promises and challenges of translational re- therapeutic proteins and peptides. Google Patents.
search. Drug Deliv. Transl. Res. 2, 238253. Rawat, M.K., Jain, A., Singh, S., 2011. Studies on binary lipid matrix based solid lipid
Mobley, W.C., Schreier, H., 1994. Phase transition temperature reduction and glass for- nanoparticles of repaglinide: in vitro and in vivo evaluation. J. Pharm. Sci. 100,
mation in dehydroprotected lyophilized liposomes. J. Control. Release 31, 7387. 23662378.
Montenegro, L., Campisi, A., Sarpietro, M.G., Carbone, C., Acquaviva, R., Raciti, G., Reverchon, E., Porta, G.D., 2001. Supercritical uids-assisted micronization techniques.
Puglisi, G., 2011a. In vitro evaluation of idebenone-loaded solid lipid nanoparticles Low-impact routes for particle production. Pure Appl. Chem. 73 (8), 12931297.
for drug delivery to the brain. Drug Dev. Ind. Pharm. 37, 737746. Ricci, M., Puglia, C., Bonina, F., Giovanni, C.D., Giovagnoli, S., Rossi, C., 2005. Evaluation
Muchow, M., Maincent, P., Mller, R.H., 2008. Lipid nanoparticles with a solid matrix of indomethacin percutaneous absorption from nanostructured lipid carriers (NLC):
(SLN, NLC, LDC) for oral drug delivery. Drug Dev. Ind. Pharm. 34, 13941405. in vitro and in vivo studies. J. Pharm. Sci. 94, 11491159.
Mudshinge, S.R., Deore, A.B., Patil, S., Bhalgat, C.M., 2011. Nanoparticles: emerging Ruktanonchai, U., Bejrapha, P., Sakulkhu, U., Opanasopit, P., Bunyapraphatsara, N.,
carriers for drug delivery. Saudi Pharm. J. 19, 129141. Junyaprasert, V., Puttipipatkhachorn, S., 2009. Physicochemical characteristics, cy-
Muller, R.H., Mader, K., Gohla, S., 2000. Solid lipid nanoparticles (SLN) for controlled totoxicity, and antioxidant activity of three lipid nanoparticulate formulations of
drug deliverya review of the state of the art. Eur. J. Pharm. Biopharm. 50, 161177. alpha-lipoic acid. Aaps Pharmscitech 10, 227234.
Mller, R., Maaben, S., Weyhers, H., Mehnert, W., 1996. Phagocytic uptake and cyto- Salmaso, S., Elvassore, N., Bertucco, A., Caliceti, P., 2009. Production of solid lipid
toxicity of solid lipid nanoparticles (SLN) sterically stabilized with poloxamine 908 submicron particles for protein delivery using a novel supercritical gasassisted
and poloxamer 407. J. Drug Target. 4, 161170. melting atomization process. J. Pharm. Sci. 98 (2), 640650.
Mller, R., Maassen, S., Schwarz, C., Mehnert, W., 1997. Solid lipid nanoparticles (SLN) Sanad, R.A., Abdelmalak, N.S., Badawi, A.A., 2010. Formulation of a novel oxybenzone-
as potential carrier for human use: interaction with human granulocytes. J. Control. loaded nanostructured lipid carriers (NLCs). Aaps Pharmscitech 11, 16841694.
Release 47, 261269. Sanjula, B., Shah, F.M., Javed, A., Alka, A., 2009. Eect of poloxamer 188 on lymphatic
Mller, R., Runge, S., Ravelli, V., Mehnert, W., Thnemann, A., Souto, E., 2006. Oral uptake of carvedilol-loaded solid lipid nanoparticles for bioavailability enhancement.
bioavailability of cyclosporine: solid lipid nanoparticles (SLN) versus drug nano- J. Drug Target. 17, 249256.
crystals. Int. J. Pharm. 317, 8289. Sarmento, B., Martins, S., Ferreira, D., Souto, E.B., 2007. Oral insulin delivery by means of
Mller, R., Runge, S., Ravelli, V., Thnemann, A., Mehnert, W., Souto, E., 2008. solid lipid nanoparticles. Int. J. Nanomed. 2, 743.
Cyclosporine-loaded solid lipid nanoparticles (SLN): druglipid physicochemical Sawant, K.K., Varia, J.K., Dodiya, S.S., 2008. Cyclosporine a loaded solid lipid nano-
interactions and characterization of drug incorporation. Eur. J. Pharm. Biopharm. 68, particles: optimization of formulation, process variable and characterization. Curr.
535544. Drug Deliv. 5, 6469.
Mller, R.H., Radtke, M., Wissing, S.A., 2002. Solid lipid nanoparticles (SLN) and na- Schfer-Korting, M., 2010. Drug Delivery. Springer, Berlin, Heidelberg.
nostructured lipid carriers (NLC) in cosmetic and dermatological preparations. Adv. Schubert, M., Mller-Goymann, C., 2003. Solvent injection as a new approach for man-
Drug Deliv. Rev. 54, S131S155. ufacturing lipid nanoparticlesevaluation of the method and process parameters. Eur.
Mumper Russell John, Michael Jay, 2006. Microemulsions as Precursors to Solid J. Pharm. Biopharm. 55, 125131.
Nanoparticles. U.S. Patent No. 7,153,525. 26 Dec. Schwarz, C., Mehnert, W., 1997. Freeze-drying of drug-free and drug-loaded solid lipid
Nam, S.-H., Ji, X.Y., Park, J.-S., 2011. Investigation of tacrolimus loaded nanostructured nanoparticles (SLN). Int. J. Pharm. 157, 171179.
lipid carriers for topical drug delivery. Bull. Korean Chem. Soc. 32, 956960. Schwarz, C., Mehnert, W., Lucks, J., Mller, R., 1994. Solid lipid nanoparticles (SLN) for
Nayak, A.P., Tiyaboonchai, W., Patankar, S., Madhusudhan, B., Souto, E.B., 2010. controlled drug delivery. I. Production, characterization and sterilization. J. Control.
Curcuminoids-loaded lipid nanoparticles: novel approach towards malaria treatment. Release 30, 8396.
Colloids Surf. B: Biointerfaces 81, 263273. Seetapan, N., Bejrapha, P., Srinuanchai, W., Ruktanonchai, U.R., 2010. Rheological and
Obeidat, W.M., Schwabe, K., Mller, R.H., Keck, C.M., 2010. Preservation of nanos- morphological characterizations on physical stability of gamma-oryzanol-loaded
tructured lipid carriers (NLC). Eur. J. Pharm. Biopharm. 76, 5667. solid lipid nanoparticles (SLNs). Micron 41, 5158.
O'driscoll, C., Grin, B., 2008. Biopharmaceutical challenges associated with drugs with Shah, R., Eldridge, D., Palombo, E., Harding, I., 2015. Lipid Nanoparticles: Production,

55
P. Ganesan, D. Narayanasamy Sustainable Chemistry and Pharmacy 6 (2017) 3756

Characterization and Stability. Springer International Publishing, USA. emulsiers: pharmacokinetic and behavioral evaluations. J. Pharm. Sci. 100,
Shahgaldian, P., Da Silva, E., Coleman, A.W., Rather, B., Zaworotko, M.J., 2003a. Para- 547557.
acyl-calix-arene based solid lipid nanoparticles (SLNs): a detailed study of prepara- ner, M., Yener, G., 2007. Importance of solid lipid nanoparticles (SLN) in various ad-
tion and stability parameters. Int. J. Pharm. 253, 2338. ministration routes and future perspectives. Int. J. Nanomed. 2, 289.
Shahgaldian, P., Gualbert, J., A Ssa, K.S., Coleman, A.W., 2003b. A study of the freeze- Unruh, T., Bunjes, H., Westesen, K., Koch, M., 2001. Investigations on the melting be-
drying conditions of calixarene based solid lipid nanoparticles. Eur. J. Pharm. haviour of triglyceride nanoparticles. Colloid Polym. Sci. 279, 398403.
Biopharm. 55, 181184. Varshosaz, J., Minayian, M., Moazen, E., 2010a. Enhancement of oral bioavailability of
Shahgaldian, P., Quattrocchi, L., Gualbert, J., Coleman, A.W., Gorelo, P., 2003c. AFM pentoxifylline by solid lipid nanoparticles. J. Liposome Res. 20, 115123.
imaging of calixarene based solid lipid nanoparticles in gel matrices. Eur. J. Pharm. Varshosaz, J., Tabbakhian, M., Mohammadi, M.Y., 2010b. Formulation and optimization
Biopharm. 55, 107113. of solid lipid nanoparticles of buspirone HCl for enhancement of its oral bioavail-
Shiau, Y., 1990. Mechanism of intestinal fatty acid uptake in the rat: the role of an acidic ability. J. Liposome Res. 20, 286296.
microclimate. J. Physiol. 421, 463. Vezz, K., Borin, D., Bertucco, A., Bersani, S., Salmaso, S., Caliceti, P., 2010. Production of
Shulkin, P.M., Seltzer, S.E., Davis, M.A., Adams, D.F., 1984. Lyophilized liposomes: a new lipid microparticles containing bioactive molecules functionalized with PEG. J.
method for long-term vesicular storage. J. Microencapsul. 1, 7380. Supercrit. Fluids 54 (3), 328334.
Silva, A., Santos, D., Ferreira, D., Souto, F., 2007. Oral delivery of drugs by means of solid Voutchkova-Kostal, A.M., Kostal, J., Connors, K.A., Brooks, Bryan W., Anastas, P.T.,
lipid nanoparticles. Minerva Biotecnol. 19, 1. Zimmerman, J.B., 2012. Towards rational molecular design for reduced chronic
Silva, A., Gonzlez-Mira, E., Garca, M., Egea, M., Fonseca, J., Silva, R., Santos, D., Souto, aquatic toxicity. Green. Chem. 14, 10011008.
E., Ferreira, D., 2011. Preparation, characterization and biocompatibility studies on Waghmare, A., Grampurohit, N., Gadhave, M., Gaikwad, D., Jadhav, S., 2012. Solid lipid
risperidone-loaded solid lipid nanoparticles (SLN): high pressure homogenization nanoparticles: a promising drug delivery. Int. Res. J. Pharm. 3 (4), 100107.
versus ultrasound. Colloids Surf. B: Biointerfaces 86, 158165. Wang, G., Wang, J., Wu, W., Tony To, S.S., Zhao, H., Wang, J., 2015. Advances in lipid-
Sjstrm, B., Bergensthl, B., 1992. Preparation of submicron drug particles in lecithin- based drug delivery: enhancing eciency for hydrophobic drugs. Expert Opin. Drug
stabilized o/w emulsions I. Model studies of the precipitation of cholesteryl acetate. Deliv. 12, 14751499.
Int. J. Pharm. 88, 5362. Westesen, K., Bunjes, H., Koch, M., 1997. Physicochemical characterization of lipid na-
Souto, E., Muller, R., 2006. Application of lipid nanoparticles (SLN and NLC) in food noparticles and evaluation of their drug loading capacity and sustained release po-
industry. J. Food Technol. 4, 9095. tential. J. Control. Release 48, 223236.
Souto, E., Wissing, S., Barbosa, C., Mller, R., 2004. Evaluation of the physical stability of Wissing, S., Kayser, O., Mller, R., 2004. Solid lipid nanoparticles for parenteral drug
SLN and NLC before and after incorporation into hydrogel formulations. Eur. J. delivery. Adv. Drug Deliv. Rev. 56, 12571272.
Pharm. Biopharm. 58, 8390. Wissing, S.A., Mller, R.H., 2003. The inuence of solid lipid nanoparticles on skin hy-
Souto, E.B., Muller, R., 2007. Lipid nanoparticles (solid lipid nanoparticles and nanos- dration and viscoelasticityin vivo study. Eur. J. Pharm. Biopharm. 56, 6772.
tructured lipid carriers) for cosmetic, dermal, and transdermal applications. DRUGS Wu, L., Zhang, J., Watanabe, W., 2011. Physical and chemical stability of drug nano-
Pharm. Sci. 166, 213. particles. Adv. Drug Deliv. Rev. 63, 456469.
Souto, E.B., Mller, R.H., 2010. Lipid nanoparticles: eect on bioavailability and phar- Xie, S., Pan, B., Wang, M., Zhu, L., Wang, F., Dong, Z., Wang, X., Zhou, W., 2010.
macokinetic changes. In: Schfer-Korting, M. (Ed.), Drug Delivery. Springer Berlin Formulation, characterization and pharmacokinetics of praziquantel-loaded hydro-
Heidelberg, Berlin, Heidelberg, pp. 115141. genated castor oil solid lipid nanoparticles. Nanomedicine 5, 693701.
Souza, L., Silva, E., Martins, A., Mota, M., Braga, R., Lima, E., Valadares, M., Taveira, S., Xie, S., Zhu, L., Dong, Z., Wang, X., Wang, Y., Li, X., Zhou, W., 2011. Preparation,
Marreto, R., 2011. Development of topotecan loaded lipid nanoparticles for chemical characterization and pharmacokinetics of enrooxacin-loaded solid lipid nano-
stabilization and prolonged release. Eur. J. Pharm. Biopharm. 79, 189196. particles: inuences of fatty acids. Colloids Surf. B: Biointerfaces 83, 382387.
tecov, J., Mehnert, W., Blaschke, T., Kleuser, B., Sivaramakrishnan, R., Zouboulis, C.C., Xu, X.-M., Wang, Y.-S., Chen, R.-Y., Feng, C.-L., Yao, F., Tong, S.-S., Wang, L., Yamashita,
Seltmann, H., Korting, H.C., Kramer, K.D., Schfer-Korting, M., 2007. Cyproterone F., Yu, J.-N., 2011. Formulation and pharmacokinetic evaluation of tetracycline-
acetate loading to lipid nanoparticles for topical acne treatment: particle character- loaded solid lipid nanoparticles for subcutaneous injection in mice. Chem. Pharm.
isation and skin uptake. Pharm. Res. 24, 9911000. Bull. 59, 260265.
Strauss, G., Schurtenberger, P., Hauser, H., 1986. The interaction of saccharides with lipid Yadav, N., Khatak, S., Sara, U.V.S., 2013. Solid lipid nanoparticlesa review. Int. J. Appl.
bilayer vesicles: stabilization during freeze-thawing and freeze-drying. Biochim. Pharm. 5, 818.
Biophys. Acta (BBA)-Biomembr. 858, 169180. Yang, R., Gao, R., Li, F., He, H., Tang, X., 2011. The inuence of lipid characteristics on
Subedi, R.K., Kang, K.W., Choi, H.-K., 2009. Preparation and characterization of solid the formation, in vitro release, and in vivo absorption of protein-loaded SLN prepared
lipid nanoparticles loaded with doxorubicin. Eur. J. Pharm. Sci. 37, 508513. by the double emulsion process. Drug Dev. Ind. Pharm. 37, 139148.
Suresh, G., Manjunath, K., Venkateswarlu, V., Satyanarayana, V., 2007. Preparation, Yang, S., Zhu, J., Lu, Y., Liang, B., Yang, C., 1999. Body distribution of camptothecin solid
characterization, and in vitro and in vivo evaluation of lovastatin solid lipid nano- lipid nanoparticles after oral administration. Pharm. Res. 16, 751757.
particles. Aaps Pharmscitech 8, E162E170. Yuan, H., Chen, J., Du, Y.-Z., Hu, F.-Q., Zeng, S., Zhao, H.-L., 2007a. Studies on oral
Svilenov, H., Tzachev, C., 2014. Solid lipid nanoparticlesa promising drug delivery absorption of stearic acid SLN by a novel uorometric method. Colloids Surf. B:
system. Nanomed. One Cent. Press Manch. 187237. Biointerfaces 58, 157164.
Swathi, G., Prasanthi, N., Manikiran, S., Ramarao, N., 2012. Solid lipid nanoparticles: Yuan, H., Wang, L.-L., Du, Y.-Z., You, J., Hu, F.-Q., Zeng, S., 2007b. Preparation and
colloidal carrier systems for drug delivery. ChemInform 43 (no). characteristics of nanostructured lipid carriers for control-releasing progesterone by
Swathi, M.P., Bala, P., 2013. A review on solid lipid nanoparticles. Int. J. Pharm. Sci. Rev. melt-emulsication. Colloids Surf. B: Biointerfaces 60, 174179.
Res 20 (2), 196206 (36). Zara, G.P., Bargoni, A., Cavalli, R., Fundar, A., Vighetto, D., Gasco, M.R., 2002.
Tan, S., Billa, N., Roberts, C., Burley, J., 2010. Surfactant eects on the physical char- Pharmacokinetics and tissue distribution of idarubicinloaded solid lipid nano-
acteristics of amphotericin B-containing nanostructured lipid carriers. Colloids Surf. particles after duodenal administration to rats. J. Pharm. Sci. 91, 13241333.
A: Physicochem. Eng. Asp. 372, 7379. Zhang, N., Ping, Q., Huang, G., Xu, W., Cheng, Y., Han, X., 2006. Lectin-modied solid
Taratula, O., Kuzmov, A., Shah, M., Garbuzenko, O.B., Minko, T., 2013. Nanostructured lipid nanoparticles as carriers for oral administration of insulin. Int. J. Pharm. 327,
lipid carriers as multifunctional nanomedicine platform for pulmonary co-delivery of 153159.
anticancer drugs and siRNA. J. Control. Release 171, 349357. Zhang, T., Chen, J., Zhang, Y., Shen, Q., Pan, W., 2011. Characterization and evaluation
Teeranachaideekul, V., Souto, E.B., Mller, R.H., Junyaprasert, V.B., 2008. of nanostructured lipid carrier as a vehicle for oral delivery of etoposide. Eur. J.
Physicochemical characterization and in vitro release studies of ascorbyl palmitate- Pharm. Sci. 43, 174179.
loaded semi-solid nanostructured lipid carriers (NLC gels). J. Microencapsul. 25, Zhang, X., Pan, W., Gan, L., Zhu, C., Gan, Y., Nie, S., 2008. Preparation of a dispersible
111120. PEGylate nanostructured lipid carriers (NLC) loaded with 10-hydroxycamptothecin
Thatipamula, R., Palem, C., Gannu, R., Mudragada, S., Yamsani, M., 2011. Formulation by spray-drying. Chem. Pharm. Bull. 56, 16451650.
and in vitro characterization of domperidone loaded solid lipid nanoparticles and Zhang, X., Liu, J., Qiao, H., Liu, H., Ni, J., Zhang, W., Shi, Y., 2010. Formulation opti-
nanostructured lipid carriers. Daru 19, 2332. mization of dihydroartemisinin nanostructured lipid carrier using response surface
Trotta, M., Debernardi, F., Caputo, O., 2003. Preparation of solid lipid nanoparticles by a methodology. Powder Technol. 197, 120128.
solvent emulsicationdiusion technique. Int. J. Pharm. 257, 153160. Zur Mhlen, A., Schwarz, C., Mehnert, W., 1998. Solid lipid nanoparticles (SLN) for
Tsai, M.J., Huang, Y.B., Wu, P.C., Fu, Y.S., Kao, Y.R., Fang, J.Y., Tsai, Y.H., 2011. Oral controlled drug deliverydrug release and release mechanism. Eur. J. Pharm.
apomorphine delivery from solid lipid nanoparticles with dierent monostearate Biopharm. 45, 149155.

56

Você também pode gostar