Sunteți pe pagina 1din 18

European Journal of Pharmaceutics and Biopharmaceutics 87 (2014) 1–18

Contents lists available at ScienceDirect

European Journal of Pharmaceutics and Biopharmaceutics


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

Review Article

Nanotoxicology applied to solid lipid nanoparticles and nanostructured


lipid carriers – A systematic review of in vitro data
Slavomira Doktorovova a,b, Eliana B. Souto b,c, Amélia M. Silva a,d,⇑
a
Department of Biology and Environment, School of Life and Environmental Sciences (ECVA, UTAD), University of Trás-os-Montes and Alto Douro, Vila Real, Portugal
b
Institute of Biotechnology and Bioengineering, Centre of Genomics and Biotechnology, University of Trás-os-Montes and Alto Douro (IBB/CGB-UTAD), Vila-Real, Portugal
c
Department of Pharmaceutical Technology, Fernando Pessoa University, UFP, Porto, Portugal
d
Centre for the Research and Technology of Agro-Environmental and Biological Sciences, University of Trás-os-Montes and Alto Douro (CITAB-UTAD), Vila-Real, Portugal

a r t i c l e i n f o a b s t r a c t

Article history: Solid lipid nanoparticles (SLN) and nanostructured lipid carriers (NLC) were developed as alternative to
Received 29 June 2013 other colloidal carriers. They were designed to overcome lipid nanoemulsions and liposomes in stability
Accepted in revised form 4 February 2014 and ability to control the release of an encapsulated substance, and at the same time to be better toler-
Available online 12 February 2014
ated than polymeric nanoparticles. Since the patenting of SLN discovery, large amount of data became
available on the behaviour of these systems in vitro. SLN/NLC have many prerequisites to be a well tol-
Keywords: erated carrier – the currently available data seem to confirm it, but there are also some contradictory
Lipid nanoparticles
results. In this review, we collected the available data from cytotoxicity, oxidative stress and hemocom-
Solid lipid nanoparticles
Nanostructured lipid carriers
patibility studies in vitro and analysed their outcomes. We also provide a summary of the available data
Cytotoxicity in a form of reference table.
Genotoxicity Ó 2014 Elsevier B.V. All rights reserved.
Oxidative stress
Blood compatibility
In vitro studies

1. Introduction at room and body temperature [3], are 2nd generation of solid li-
pid-based colloidal carriers, with improved stability and drug
Solid lipid nanoparticles (SLN), nanoparticles prepared from a encapsulation ability. SLN and NLC have all pre-requisites to enter
lipid matrix that is solid at body and room temperature, stabilised the market faster than other colloidal carriers – among the reasons
by suitable surfactants and having size <1 lm, were invented it is the use of starting materials that are biodegradable and al-
20 years ago and, since then they have received great and still ready in use in pharmaceutical and cosmetic products, many of
increasing attention in pharmaceutical technology research [1,2]. them of GRAS status (generally regarded as safe). Several cosmetic
Nanostructured lipid carriers (NLC), i.e. nanoparticles composed products based on SLN are already marketed [4]. Secondly, provid-
of a mixture of a solid and a liquid lipid which lipid matrix is solid ing an advantage over liposomes or polymeric nanoparticles, prep-
aration methods feasible at industrial scale are available, and these
Abbreviations: Chol, cholesterol; COM, Compritol™ 888 ATO; CP, cetyl palmitate; methods avoid the use of organic solvents [5]. Furthermore, in vitro
CPC, cetylpyrimidium chloride; CPT, camptothecin; CTAB, cetyltrimethylammonium
tolerability of SLN and NLC appears to be much higher than that of
bromide, hexadecyltrimethylammonium bromide; CysA, Cyclosporine A; DOPE,
dioleoylphosphatidyl ethanolamine; DOTAP, N-[1-(2,3-dioleoyloxy)propyl]-N,N,N- polymeric nanoparticles (see further).
trimethylammonium chloride; DOX, doxorubicin; DTX, docetaxel; EQ1, esterquat1; Toxicology of nanomaterials is becoming an important issue
EYPS, egg yolk phosphatidyl choline; FA, fatty acid(s); GMS, glyceryl monostearate; nowadays, especially with regard to nanomaterials present in envi-
GO, glyceride oleate; MTX, methotrexate; OA, oleic acid; P188, Poloxamer 188; P407,
ronment and nanomaterials intended for medical use. The biggest
Poloxamer 407; PEG, polyethyleneglycol; p-gp, p-glycoprotein; PTC, paclitaxel; PVA,
polyvinylalcohol; SA, stearic acid; SDS, sodium dodecyl sulphate; StA, stearylamine; concern about nanomaterial safety is focused on nanomaterials with
STC, sodium taurocholate; T80, Tween 80; TL, glyceryl trilaurate, trilaurine; TM, at least one dimension smaller than 100 nm. Although SLN typically
glyceryl trimyristate, trimyristine; TP, glyceryl tripalmitate, tripalmitine; TS, glyceryl do not reach this size (z-average data are most often reported over
tristearate, tristearine; +, lipids (or surfactants) used simultaneously in one lipid 100 nm; it was suggested SLN be called ‘‘submicron particles’’ rather
matrix; /, lipids (or surfactants) used in the same study in different lipid matrices.
⇑ Corresponding author at: Department of Biology and Environment, School of than nanoparticles [6]), facilitated cellular uptake of lipid nanoparti-
Life and Environmental Sciences (ECVA, UTAD), University of Trás-os-Montes and cles themselves or their drug payload have been reported repeatedly.
Alto Douro, Quinta de Prados, P-5001-801 Vila Real, Portugal. Tel.: +351 SLN and NLC have many pre-requisites to be safe nanocarriers.
259350106; fax: +351 259350480. Indeed during the first 10 years of development very encouraging
E-mail address: amsilva@utad.pt (A.M. Silva).

http://dx.doi.org/10.1016/j.ejpb.2014.02.005
0939-6411/Ó 2014 Elsevier B.V. All rights reserved.
2 S. Doktorovova et al. / European Journal of Pharmaceutics and Biopharmaceutics 87 (2014) 1–18

results were reported. However, for approval in use in medicinal order to calculate IC50, several concentrations need to be tested. If
products, more evidence will be required. Research reports reflect IC50 is not required for further studies, many researchers report a
this challenge, even despite expected low toxicity or no toxicity at concentration at which no toxicity is observed (or toxicity at
all. Up to date, considerable amount of data on SLN/NLC behaviour acceptable levels) and select this concentration for further studies.
in cell culture is available. As of December 2013 (accessed on 1st of This reporting is naturally less precise. Decrease in cell viability by
December 2013), a Pubmed search by keywords ‘‘solid lipid nano- more than 30% (i.e. cell viability of 70% and less) is to be considered
particles’’ and ‘‘cytotoxicity’’ returns 128 results; the same search as a cytotoxic effect [16]. This norm is frequently forgotten when
in ISI Web of Knowledge returns 174 entries. The existing data on referring the results, thus it is important to review the actual cell
toxicity in cell culture experiments are however still difficult to viability % reported rather than the evaluation made by the authors.
compare – they were obtained (i) in various cell lines and cell types, Besides cell viability studies, the advanced toxicological studies
at different passages, (ii) the methods used have slight variations also search for effects on cells that do not necessarily lead to cell
among research groups, and, clearly, (iii) different formulations death. These include oxidative stress, i.e. increased production of
for various purposes were tested. To the best of our knowledge, a reactive oxygen species (ROS); lipid (integral part of cell mem-
systematic study with a large series of formulations and performed branes) peroxidation, imbalance in oxidised/reduced glutathione
in one sole laboratory has not been published up to date. pool or DNA damage. An excellent revision of toxicology of nanom-
Among the currently available data, there are a few studies aterials including various methods was published by Lewinski and
which main purpose is to address the safety issues of SLN/NLC collaborators [17] where more detail on the toxicological assays
for the selected route of administration: Weyenberg and collabora- can be obtained.
tors tested a series of SLN with both positive and negative surface
charge for the purpose of dermal delivery [7], Ridolfi and collabo-
rators [8] also tested a small series of SLN in fibroblasts and kerat- 2.1. Cell viability assays
inocytes. Another group tested two SLN formulations intended for
lung delivery by different cell viability assessment methods and The majority of cell viability studies of SLN/NLC were performed
also reports an ex vivo study in murine lung tissue [9,10]. Tolerabil- by MTT assay. MTT is a tetrazolium dye which is converted into
ity of a series of SLN by normal human granulocytes from healthy formazan by metabolically active cells [18]. The product is insolu-
volunteers and leukemic HL-60 macrophages was tested by the ble in water; a solubilisation step by an organic solvent is neces-
group of Müller [11–13]. Petersen and colleagues [14] investigated sary to dissolve the crystals and enable absorbance reading. The
the influence of lipid physical state on the toxicity towards cells in use of an organic solvent may raise the logical question if the lipids
culture and Silva and collaborators [15] reported a study with a used for SLN may be solubilised as well. Even though some lipids
series of SLN in monkey kidney cells. such as Precirol ATO 5 (Glycerol distearate, Gattefosse SAS) are sol-
Cell studies have also been performed for various purposes. uble in chloroform [19], the extent of solubilisation of the particles
Studies using chemotherapeutic drug(s) loaded in SLN/NLC usually should be low. Furthermore, SLN/NLC in the dispersion themselves
include a cell viability study to prove that the formulation is absorb visible light. Absorbance reading of MTT assay is usually
efficient. The main reason of this testing is a proof of efficiency of done at 570 nm; at this wavelength the absorbance from SLN/
the drug loaded formulation. A control using the blank SLN/NLC is NLC is very low. To avoid the possibility of lipid interaction with
a logical component of such a study to prove that the carrier itself the assay, some research groups include a washing step before
is not toxic, however, there are reports where this control is omitted incubation with organic solvent [7,20,21]. Again, majority of pub-
and others that only refer to blank SLN/NLC non-toxicity marginally. lished reports do not enclose if the particles had been removed
Improvement in chemotherapeutic drug delivery is a hot topic in or not before incubation with MTT.
current research; SLN/NLC research also follows this trend and there Apart from MTT, other formazan dye based assays have been
are up to 100 formulations reported, most of them with cell toxicity used, including Alamar Blue (AB), WST-1 and WST-8 (Water Solu-
data. These are useful sources of information on SLN/NLC safety. ble Tetrazolium salts) based assays. These dyes are water-soluble;
Another big source of cell viability data are SLN/NLC formulations the absorbance reading can be done without solubilisation step.
intended for gene delivery. Again, the proof of efficiency requires the When using these assays, the presence of SLN in the cell culture
use of cell culture and the information of the maximal tolerable con- media is even more critical. The blank measurements should in-
centration that can be used in transfection experiments. Furthermore, clude also the particles in all concentrations used in the experi-
lately more and more reports on non-chemotherapeutic drugs stud- ment. Fig. 1 shows such blanks and illustrates that over a
ies include cell viability assays; aiming to show that neither the blank
nor the drug loaded formulation causes cellular damage/toxicity.
In order to compare the published results, we collected the
information on cell lines, methods used, time of exposure, concen-
tration range and the study outcome, together with basic informa-
tion about the formulations that were tested (solid lipid, surfactant,
size – mostly reported as z-average diameter, zeta potential – ZP).
Outcomes of the studies are reported here as given by the authors;
i.e., by % of viability at a particular concentration or as IC50. Concen-
tration of SLN/NLC in the tested dispersion is given as mass solid
lipid per volume, when possible to calculate this information from
the published data. For uniformity, the concentrations are all given
here in mg/ml, i.e., solid lipid content/ml cell culture media.

Fig. 1. Absorbance at 570 nm of various SLN in DMEM. Absorbance readings were


obtained from Multiskan Ex microplate reader (MTX Labsystems, USA) and are
2. Methods for risk identification in vitro
presented as mean from quadruplicate with standard deviation. (A, B and C)
Represents three distinct SLN formulations, differing among themselves in solid lipid
Basic test for a toxicological study is a cell viability study. The and/or surfactant composition. Unpublished data (SD and AMS), for information on
endpoint is a concentration that causes 50% cell death (IC50). In formulation composition, please see Doktorovova, et al. 420 (2011) 341–349.
S. Doktorovova et al. / European Journal of Pharmaceutics and Biopharmaceutics 87 (2014) 1–18 3

concentration range the blank absorbance shows remarkable dif- 3. Special issues with SLN
ferences, depending on the SLN composition.
Other type of cell viability assay is based on dye uptake. Neutral By definition, SLN/NLC are formulated in aqueous dispersion;
red (NR), Crystal violet or Trypan Blue were used in SLN/NLC tests. higher concentration of electrolytes may cause instability and pre-
NR assay is based on accumulation of NR in lysosomes of living cipitation of SLN [27]. This is an especially important issue in cell
cells [22]; Trypan Blue is based on the fact that living cells can ac- studies with this kind of carriers, as they need to be performed
tively exclude the dye, while dead cells accumulate the dye [23]. in buffers (to maintain physiological pH and osmolarity) or cell cul-
The evaluation is based on the count of living cells vs. dead cells ture media. This fact should immediately raise a concern about
marked by the accumulated dye. SLN/NLC stability in these media. The vast majority of reports do
Serving for the same purpose (estimation of % of living cells) but not address this issue at all. From the few reports that include
based on different mechanism; these two types of methods are some stability data we can conclude that SLN/NLC can be formu-
complementary and were used simultaneously within one study lated so that they were stable in these media.
with the same materials and cell lines [9,15]. From these two re- Petersen and collaborators [14] reported no aggregation of their
ports it can be concluded that the results are comparable: the re- samples in DMEM at 37 °C in the whole range of concentrations
sults of Silva and collaborators showed excellent correlation of used in cell studies, Silva and colleagues [28] also reported that
the results obtained with MTT and NR results in MDCK cells; a the tested SLN were stable in cell culture media, although at lower
slightly higher IC50 was calculated by NR assay in Vero cells [15]. concentrations. Almost no change in size over a large concentra-
Nassimi and collaborators [9] reported an IC50 for the tested SLN tion range (0.9 ng/ml–9 mg/ml) in a buffer was reported by Rivolta
of 2.09 mg/ml by NR assay and of 3.09 mg/ml by MTT assay. These and collaborators [29], How and collaborators [30] reported that
findings are perfectly in agreement with previous comparisons of their NLC formulations were stable in RPMI with or without foetal
various cell viability assays [24]. bovine serum (FBS), showing only few nm difference from the ori-
Lactate dehydrogenase (LDH) release assay can also be taken as ginal z-ave value in aqueous dispersion. This change occurred
a measure of cell viability. It reflects the damage to cell membrane, within 60 min of incubation; up to 5 h no further changes in diam-
thus gives additional information to mitochondrial activity-based eter were observed [30]. On the other hand, Siddiqui and collabo-
assays. Some authors, however, reported that LDH assay is less sen- rators [31] reported aggregation of cationic SLN in cell culture
sitive to cell damage than MTT or WST assays (see further). media. Positively charged SLN are usually less stable then nega-
The results that we mention here were collected at time points tively charged SLN, hence this is an expected result. Some aggrega-
varying from 4 h through the most common 24 h, 48 h to 72 h tion that could be prevented by ultrasound treatment was also
incubation times. This fact causes even further difficulties in com- mentioned for negatively charged SLN [28,32].
paring them. Ma and colleagues showed that MTT metabolism by Some authors adjusted their formulations to physiological pH
HaCaT cells shows a decrease between 4 h and 12 h of incubation. before all further tests took place [33–36], and others worked with
After 12 h, the MTT conversion gradually increased again [25]. On freeze-dried SLN/NLC that were directly re-dispersed in appropri-
the contrary, Zhuang and collaborators [26] observed steady de- ate buffer solution [37]. In response to the need of stable
crease in IC50 of three drug-loaded SLN formulations (mitoxan- SLN/NLC dispersions for in vitro studies, Peters and collaborators
throne, methotrexate, paclitaxel; using MCF-7 cell line) [38] prepared NLC especially with the aim of being stable in buffer
determined at 24 h, 48 h and 72 h. solutions.

Box 1 – List of substances used in cell viability assays.


4. SLN effect on cells in culture

MTT 3-(4,5-dimethyltiazol-2-yl)-2,5-diphenyl 4.1. Comparison by IC50


tetrazolium bromide
MTS 3-(4,5-dimethyltiazol-2-yl)-5-(3- The most accurate comparison of SLN/NLC toxicity can be done
carboxymethoxyphenyl)-2-(4-sulfophenyl)- by using the IC50 values provided by the authors. Although very
dihydro tetrazolium few studies were conducted with enough concentrations to calcu-
XTT 2,3-bis-(2-methoxy-4-nitro-5sulfophenyl)- late IC50, all of them tested a set of formulations which allows look-
dihydroxy-terazolium-5-carboxyanilide ing at the overall information. Table 1 lists these results from the
WST-1 2-(4-Iodophenyl)-3-(4-nitrophenyl)-5-(2,4- most toxic (lowest IC50) to the most well tolerated (highest IC50).
disulfophenyl)-2H-tetrazolium From a brief look it is apparent that the IC50s are found mostly in
WST-8 2-(2-methoxy-4-nitrophenyl)-3-(4- the range of 0.1–1.0 mg/ml particles in dispersion. Higher IC50s
nitrophenyl)-5-(2,4-disulfophenyl)- than 1 mg/ml were reported by several authors, e.g. Silva and col-
dihydroxy tetrazolium laborators [15] reported an IC50 over 1 mg/ml, Schubert and Mul-
Alamar Blue 7-hydroxy-3-hydro-phenoxazin-3-one-10- ler-Goymann [39] reported a value of 1.3 mg/ml, or Nassimi and
(AB) oxide), syn. Resazurin collaborators [9] 2–3 mg/ml.
Crystal violet Tris(4-(dimethylamino)phenyl)methylium There is no clear tendency of neither surfactant nor lipid core
chloride, syn. Gentian violet influence on the obtained IC50 values. We can find glyceryl monos-
Neutral Red 3-amino-7-dimethylamino-2- tearate (GMS)-composed formulations in both ends of the table,
(NR) methylphenazine hydrochloride, syn. including both GMS-alone and in combination with stearic acid
toluylene red (SA) and oleic acid (OA). Surfactants are considered to be decisive
Propidium 3,8-diamino-5-[3- in toxicity of nanoparticles [40], from the listed IC50 (Table 1) it
Iodide (PI) (diethylmethylammonio)propyl]-6- can be concluded that only poly-vinyl alcohol (PVA) and sodium
phenylphenanthridinium diiodide dodecyl sulphate (SDS) used as surfactants lead to low IC50 values,
Trypan Blue (3Z,30 Z)-3,30 -[(3,30 -dimethylbiphenyl-4,40 - which is an expected finding, and that frequently used surfactants
diyl)di(1Z)hydrazin-2-yl-1-ylidene]bis(5- in SLN/NLC formulations like Tween 80 (T80) are to be find in the
amino-4-oxo-3,4-dihydronaphthalene-2,7- bottom of the table.
disulfonic acid), syn. Azidine Blue With exact measure of toxicity as IC50, it is possible to compare
the influence of size and charge. The importance of size on toxicity
4 S. Doktorovova et al. / European Journal of Pharmaceutics and Biopharmaceutics 87 (2014) 1–18

Table 1
Effect of blank SLN and NLC (i.e. without any encapsulated active ingredient) on cell viability expressed as half maximal inhibitory concentration (IC50). The results are ordered
from the most toxic formulations (lowest IC50) to formulations with lowest effect on cell viability (highest IC50). The column ‘‘drug’’ refers to the substance that the formulation
was intended to encapsulate.

Type Lipid matrix composition Surfactant Drug Size; PdI; ZP Cell line Assay Exposure IC50 (mg/ml) Refs.
t (h) [] (mg/ml)
SLN+ CP, DOTAP Oleth20, 522 nm; 0.29; n.a. Du-145 MTT 24 0.005–2.5 0.125 [43]
GO
SLN GMS Chitosan, DOX 83–164 nm; 0.15–0.23; MCF-7 MTT 48 n.a. 0.17456 [37]
PVA 18 to 22 mV
SLN(?) S154 Soy PC, n/a 145 nm, 0.15, 17.3 mV MCF-7 MTT 72 0.037–0.3 0.2223 [45]
octadecanol
SLN TS SDS n/a 116 nm; 0.22; 39 mV Vero MTT 24 0.1–1.0 0.247 ± 0.007.2 [15]
SLN(?) S154 Soy PC, n/a 145 nm, 0.15, 17.3 mV MCF-7 MTT 48 0.037–0.3 0.2621 [45]
octadecanol
SLN(?) S154 Soy PC, n/a 145 nm, 0.15, 17.3 mV MDA-MB- MTT 72 0.037–0.3 0.275 [45]
octadecanol 231
SLN(?) S154 Soy PC, n/a 145 nm, 0.15, 17.3 mV MCF-7 MTT 24 0.037–0.3 0.2875 [45]
octadecanol
SLN(?) S154 Soy PC, n/a 145 nm, 0.15, 17.3 mV MDA-MB- MTT 24; 48 0.037–0.3 0.29 [45]
octadecanol 231
SLN GMS + SA-FA PTC 300–460 nm; n.a.; n.a. A549 MTT 48 n.a. 0.30872 ± 0.04189 [57]
SLN SA P188 PTC 300–460 nm; n.a.; n.a. A549 MTT 48 n.a. 0.33112 ± 0.03412 [57]
SLN+(?) Triolein, Chololeate, Chol, DSPE-PEG (Plasmid) 102 nm; n.a.; n.a. HepG2 WST- 10 n.a. 0.3335 [127]
DC-Chol, DOPE 1
SLN GMS + SA-PEG PTC 300–460 nm; n.a.; n.a. A549 MTT 48 n.a. 0.34789 ± 0.04240 [57]
SLN GMS P188 PTC 300–460 nm; n.a.; n.a. A549 MTT 48 n.a. 0.39682 ± 0.03319 [57]
SLN GTS P188 PTC 185 nm; 0.2; 31.7 mV A549 MTT 48 n.a. 0.40149 ± 0.02768 [57]
SLN COM P188 PTC 300–460 nm; n.a.; n.a. A549 MTT 48 n.a. 0.47148 ± 0.03091 [57]
NLC GMS + OA P188 PTC, DOX 134.4 ± 21.0 nm; MCF-7 MTT 48 n.a. 0.45549 ± 0.02768 [66]
0.361 ± 0.024;
45.5 ± 3.0 mV
NLC GMS + OA P188 PTC, DOX 134.4 ± 21.0 nm; MCF-7/Adr MTT 48 n.a. 0.46974 ± 0.03002 [66]
0.361 ± 0.024;
45.5 ± 3.0 mV
SLN Solid white vaseline USP T80 n/a 128 nm; 0.32; 18 mV Vero MTT 24 0.1–1.0 0.489 ± 0.004.9 [15]
SLN GMS P188 PTC, DOX 273 nm, 0.101, 36.4 mV SKOV-3 MTT 48 n.a. 0.544 [128]
SLN GMS P188 PTC, DOX 273 nm, 0.101, 36.4 mV MCF-7 MTT 48 n.a. 0.549 [128]
SLN GMS P188 PTC, DOX 273 nm, 0.101, 36.4 mV MCF-7/ADR MTT 48 n.a. 0.564 [128]
SLN GMS P188 PTC, DOX 273 nm, 0.101, 36.4 mV Skov-3/ MTT 48 n.a. 0.558 [128]
TR30
SLN(?) S154 + PhospholiponP90G Solutol n/a 98 nm; 0.148; 14.6 mV PCSL WST- 24 n.a. 0.575 [9]
HS15 1
SLN TS SDS n/a 116 nm, 0.22, 39 mV MDCK MTT 24 0.1–1.0 0.586 ± 0.015 [15]
SLN TS SDS n/a 116 nm; 0.22; 39 mV MDCK NR 24 0.1–1.0 0.603 ± 0.0027 [15]
SLN TS SDS n/a 116 nm; 0.22; 39 mV Vero MTT 24 0.1–1.0 0.643 ± 0.0024 [15]
SLN TS T80 n/a 110 nm; 0.22; 26 mV Vero MTT 24 0.1–1.0 0.682 [15]
SLN TS T80 n/a 110 nm; 0.22; 26 mV Vero NR 24 0.1–1.0 0.694 ± 0.0093 [15]
SLN CP P60 CPT <200 nm; <0.25; 20 mV U373 MTT 24 0.005–50 0.716 [129]
SLN CP P60 CPT A172 MTT 24 0.005–50 0.863 [129]
SLN S154 + PhospholiponP90G Solutol n/a 95 nm; 0.182; 9.8 mV HDF(NHDF) 24 0.0–3.0 (0–6%) 1.3 [39]
HS15
SLN CP P80 CPT <200 nm; <0.25; 20 mV U251 MTT 24 0.005–50 1.17025 [129]
SLN CP P80 CPT <200 nm; <0.25; 20 mV A172 MTT 24 0.005–50 2.06 [129]
SLN CP P60 CPT <200 nm; <0.25; 20 mV U251 MTT 24 0.005–50 2.086 [129]
SLN S154 + PhospholiponP90G Solutol n/a 98 nm; 0.148; 14.6 mV A549 MTT 24 0–15.0 2.09 [9]
HS15
SLN CP P80 CPT <200 nm; <0.25; 20 mV U373 MTT 24 0.005–50 2.135 [129]
SLN S154 + PhospholiponP90G Solutol n/a 98 nm; 0.148; 14.6 mV A549 MTT 24 0–15.0 3.09 [9]
HS15
SLN S154 + PhospholiponP90G Solutol n/a 144 nm; 0.225; 12.3 mV/ A549 MTT 24 0–15.0 3.43 (SLN20); 1.253 [10]
HS15 93 nm; 0.157; 22.6 mV (SLN50)
SLN CP P60 or P80 CPT <200 nm; <0.25; 20 mV U87 MTT 24 0.005–50 5.0 [129]
SLN CP P60 or P80 CPT <200 nm; <0.25; 20 mV THP1 MTT 24 0.005–50 5.0 [129]
SLN Plant lipids T80 n/a n.a.; n.a.; n.a. Vero, MTT 24 0.1–1.0 IC50  1 mg/ml (100% [15]
MDCK viability at 0.5 mg/ml)
SLN Solid white vaseline USP T80 n/a 128 nm; 0.32; 18 mV MDCK MTT 24 0.1–1.0 IC50  1 mg/ml (100% [15]
viability at 0.5 mg/ml)
SLN TS T80 n/a 110 nm; 0.22; 26 mV MDCK MTT 24 0.1–1.0 IC50  1 mg/ml (100% [15]
viability at 0.5 mg/ml)
SLN Solid white vaseline USP T80 n/a 128 nm; 0.32; 18 mV Vero NR 24 0.1–1.0 IC50  1 mg/ml (100% [15]
viability at 0.5 mg/ml)
SLN Solid white vaseline USP T80 n/a 128 nm; 0.32; 18 mV MDCK NR 24 0.1–1.0 IC50  1 mg/ml (100% [15]
viability at 0.5 mg/ml)
SLN TS T80 n/a 110 nm; 0.22; 26 mV MDCK NR 24 0.1–1.0 IC50  1 mg/ml (100% [15]
viability at 0.5 mg/ml)

Abbreviations (in this table) n.a. = not available, NR = Neutral Red uptake method, MTT = MTT method; WST-1 = WST-1 method.
S. Doktorovova et al. / European Journal of Pharmaceutics and Biopharmaceutics 87 (2014) 1–18 5

has been reported for other types of colloidal carriers [41,42]. Here, Box 3 – List and identification of cancer cell lines.
with the few data available, we cannot draw any conclusion yet.
Most of the tested SLN/NLC had size below 200 nm, but among
the formulations with lowest IC50 we can find formulation with A172 human glioma cells
z-average (z-ave) of 522 nm [43]. The size effect reported for other A549 human lung carcinoma cells
colloidal carriers like carbon nanotubes, silica nanoparticles or B16F10 murine melanoma cells
other types of inorganic particles reported on substantially smaller BEL7402 human hepatoma cells
particles, i.e., 20–40 nm [44]. This means that most SLN/NLC effects bEnd.3 murine cerebral cortex endothelioma cells
on cells cannot be explained just on their ‘‘small size’’ (which com- BxPC-3 human pancreatic adenocarcinoma cells
pared to inorganic nanoparticles is nothing but small, actually). Caco-2 human colorectal adenocarcinoma cells
Fig. 2 shows graphically the influence of SLN/NLC size on cell CPH 54A human small-cell lung cancer cells
viability at 24 h time point, collected from both IC50 and cell viabil- CT26 murine colon carcinoma cells
ity data. As can be observed in this figure, no clear tendency of Du-145 human prostate cancer androgen-non-
influence of size can be traced, although the highest values of via- responsive
bility with the highest concentrations tested were observed with HCT116 human colorectal carcinoma
SLN ranging from 100 to 300 nm. HCT-15 human colorectal carcinoma
HDF(NHDF) normal human dermal fibroblasts
Box 2 – List and identification of non-cancer cell lines.
HeLa human cervical adenocarcinoma
HepG2 human hepatocellular carcinoma
293T human kidney epithelial cell line HL-60 human promyelotic (susp) acute
3T3 BALB/ murine embryonic fibroblast promyelocytic leukemia
c HNGC1 human glioma cells
TC-1 lung epithelial cells HT-29 human colorectal adenocarcinoma
A30 human alveolar cells HuH6, HuH7 human hepatoblastoma
COS-1 African green monkey kidney fibroblast-like cell J774 murine monocyte/macrophage macrophage
line (from reticulum cell sarcoma)
COS-7 African green monkey kidney fibroblast-like cells L929 murine aneuploid fibrosarcoma
HaCaT human dermal fibroblast cells K562 Human myelogenous leukemia
MDBK Madin–Darby bovine kidney LAN5 human neuroblastoma
MDCK dog kidney fibroblasts MCF-10A human breast adenocarcinoma
NiH/3T3 murine embryonic fibroblast MCF-7 human breast adenocarcinoma
NHFa human foreskin fibroblast MCF-7/Adr human breast adenocarcinoma, resistant to
NR6-W mouse fibroblast cell lineb Adriamycin
NR8383 rat alveolar macrophages MCF-7/MX human breast adenocarcinoma, multidrug
RAT-1 rat immortalised fibroblasts resistant (Mitoxanthrone)
SDHCEC human corneal epithelia cell MDA-MB-231 human breast adenocarcinoma
TC-1 lung epithelial cells MDA-MB-435 human breast ductal adenocarcinoma
U87MG human brain-microvascular endothelial cells Na 1300 murine neuroblastoma
VK2/E6E7 human vaginal epithelial NCI/ADR-RES human ovarian cancer
Vero African green monkey kidney fibroblasts OCI-AML-2 human acute myeloid leukemia
a
OVCAR-3 human ovarian adenocarcinoma
Also designed as NHDF.
b
Transfected to express the human EGF receptor.
PC-3 human prostate adenocarcinoma
RAW 264.7 murine macrophage, Abelson murine
leukemia virus transformed
SKOV-3 human ovary adenocarcinoma
SKOV-3/TR30 human ovary adenocarcinoma, paclitaxel
resistant
SW-480 human colorectal adenocarcinoma
THP1 human acute monocytic leukemia
(phagocytic)
U87 human glioblastoma; astrocytoma
U118 human glioblastoma
U251 human neuronal glioblastoma
(astrocytoma)b
U373 human glioblastoma cellsa
a
Cell line was withdrawn by ATCC.
b
Identical to U373 [111].

Fig. 2. The effect of SLN/NLC size (z-ave, diameters in nm) on cell viability in vitro.
Cell viability (in % viable cells) vs. concentration of SLN/NLC in cell culture media 4.2. Comparison by cell viability data
reported in references listed in Tables 1–3 were used. From reports that list three
and more different SLN/NLC concentration, the highest and lowest cell viabilities Much more data on SLN/NLC safety are reported as % of viable
were used. Citation, in the respective references, as ‘‘No decrease in cell viability’’
was plotted as 100% viability, as ‘‘no toxicity’’ was plotted as 85% viability (as non-
cells in the culture, or stating that no toxicity was observed at a
toxic could be between 70% and 100% viability) and IC50 data were plotted as 50% certain concentration or concentration range. From these data,
viability. Size not available on the reference denoted by n.a. when possible to gather all necessary data, we summarised
6 S. Doktorovova et al. / European Journal of Pharmaceutics and Biopharmaceutics 87 (2014) 1–18

Tables 2 and 3 assuming that viability >70% is considered as ‘‘no tox- concentrated SLN dispersion is needed in order to achieve higher
icity’’ (Table 2) and in case that viability <70% was reported the result SLN concentration in cell culture so that the amount of cell culture
is considered as ‘‘cytotoxic effects’’ were observed (Table 3). In order media did not need to be reduced [12,14].
to better understand the data, we plotted the reported cell viability Although there are some isolated reports on the lack of toxicity
or IC50 vs. SLN/NLC concentration, for 24 h exposure (Fig. 3). at higher SLN/NLC concentration, the reasonable range to design a
About 100% viability was reported up to 0.1 mg/ml SLN in hu- toxicological study with this type of system seems to be below
man granulocytes [13] or NHF-cells [46], up to 0.5 mg/ml SLN com- 1 mg/ml.
posed by cetyl esters had no influence on 3T3 cells viability [8], up
to 1 mg/ml in NHF cells [47] and 100% viability at 1.32 mg/ml was
4.3. Influence of lipid-core material
reported by Trombino and collaborators [48] in RAT-1 cells. SLN at
1 mg/ml and 2.5 mg/ml without decrease in cell viability (HL-60
In one of the very first in vitro trials with TM and COM-com-
and MCF-7 cells) was reported by Miglietta and colleagues [49].
posed SLN (stabilised by different poloxamers) it was noted that
The only ‘‘100% viability’’ report on NLC discloses 0.9 mg/ml NLC
although overall very well tolerated, COM-SLN had slightly higher
in NHF cells, tested within the same studies as SLN (0.1 mg/ml)
impact on cell viability than TM-SLN. This was attributed to the
[46]. It is interesting to note that all of these results come from
longer time needed for degradation of the longer-chain lipids by li-
studies on cell lines that do not originate from cancer cells. The
pases [13].
material used for the formulation includes common lipids used
In a further study with a series of SLN composed of different lipid
in SLN and NLC preparation – Compritol 888 ATO (COM), trimyri-
core but stabilised with same surfactant, Scholer and collaborators
state (TM), cetyl palmitate (CP), Witepsol E58 and less frequently
[56] reported significant influence of solid lipid. COM, TM, and CP
used Apifil (PEG-8 beeswax) or cetyl esters; the surfactants include
did not cause any cytotoxic effect up to 1 mg/ml, but stearic acid-
poloxamers, polysorbates (Tweens), phospholipids, sodium tauro-
composed SLN caused a drastic decrease (2.2% cell viability) at this
cholate and even butanol.
concentration. Tristearate (TS) and GMS also were cytotoxic to mac-
Over 90% viability was reported for concentrations varying from
rophages at 1.0 mg/ml (around 60% and 70% viability, respectively).
0.015 to 1.5 mg/ml in HL-60 cells (Poloxamer stabilised SLN, while
The reason seems to be attributed to stearic acid, both as lipid-
T80 SLN stabilised reduced viability to 50% at 0.1 mg/ml [13], and
matrix material and component of respective mono- and tri-
all other reports of >90% viability state exposure of 0.1–0.4 mg/ml,
glycerides [56]. Miglietta and colleagues [49] reported however
including both cancer (HeLa [50], Raw 264.7 [51]) and non-cancer
reported no decrease in cell viability when using 1 mg/ml stearic
cell lines (HaCaT [7]). Over 80% viability of various formulations
acid-SLN. The study by Scholer et al. was performed in primary
was reported, again roughly in the same concentration range,
macrophages, the study by Miglietta et al. used HL-60 macrophages
including cationic SLN formulations (stearic acid + stearylamine
[56,49]. Interestingly, primary, ‘‘normal’’ macrophages were more
in COS-1 cells [52] and GMS + LHNH in RAW 264.7 [33]).
susceptible to damage. Stearic acid is a widely used lipid in SLN
Maintaining viability >70% was in fact reported over broad con-
and NLC formulations; however cytotoxicity testing was not per-
centration ranges: 0.4–6.0 mg/ml [53] or up to 15.0 mg/ml [54].
formed in the most of the cases. From the few reports of stearic
The lowest viabilities reported come logically from reports on
acid-based SLN we can only conclude that concentrations about
cationic SLN/NLC. 0.03 mg/ml SLN composed of stearic acid and
0.1–0.2 mg/ml are mostly reported as the highest non-toxic
stearylamine were toxic to HaCaT, 3T3 and J774 cells; viability
amounts [52,57].
<70% was reported also for stearic acid-composed SLN in the same
Ridolfi and collaborators tested a series of wax-based SLN and
study, while Witepsol composed SLN were not toxic [7].
reported different cell viabilities for cetyl esters, cetyl palmitate
From these results, it might seem that cancer cell lines were
and myristyl myristate SLN, showing that cetyl palmitate-based
more susceptible to SLN/NLC than non-cancer cell lines. The same
SLN caused the lowest obtained cell viability values in the tested
tendency was noted by Martins et al. [32] who observed higher
concentration range (up to 0.5 mg/ml, cell viability >70%) [8].
toxicity of the tested SLN towards a series of glioma cells than to-
Other reports with similar sets of SLN/NLC (different lipid, same
wards monocytes. This apparent tendency may still be misleading,
surfactant) did not point out any significant differences [47,57].
as it is not possible to draw a conclusion if this is true from the
How and colleagues tested the components of their NLC formula-
available information. The reason is different endpoints of the
tion alone; in 1% acetone in RPMI mixture. In this work, tripalmi-
studies summarised on Tables 1–3. The data cited for Tables 2
tate (TP) reduced viability of BALB/c 3T3 cells to 30% only, while
and 3 are mostly taken from studies that did not focus on toxicity
trilaurine or hydrogenated palm oil were not toxic [30]. SLN/NLC
in much detail or were designed to prove that the given formula-
may form aggregates in cell culture media or suffer changes to
tion is not toxic at a given concentration. Most of the experiments
its structure (e.g., desorption of surfactants). Testing of the unpro-
with cancer cell lines studied the toxicity in much more detail and
cessed starting material present in the same quantities and ratios
provided IC50 results. The reported IC50 (=50% viability) are mostly
as in the final formulation would therefore clarify if the observed
in the range 0.1–1.0 mg/ml which is exactly the same concentra-
effects are result of that particular combination of materials per
tion range where no cytotoxic effects (>70% viability, Table 2) are
se or of the SLN/NLC in question. Controls like the one reported
reported.
by How and colleagues [30] are rare. One reason is the materials
Fig. 4 correlates the available data on cell viability vs. SLN/NLC
are already considered safe; the other is the practical point of
concentration discriminated by the type of cell line (normal vs.
testing water-insoluble solid lipids in quantities usually used in
cancer cell line). As can be seen, no tendency of toxicity suscepti-
SLN/NLC without damaging the cells with necessary organic
bility can be attributed to normal or cancer cell lines.
solvents (DMSO, acetone, or others).
An interesting explanation of why low toxicity of SLN/NLC is of-
ten reported was given by Liu and colleagues [55] – blanks SLN are
tested in concentrations suitable to deliver small amounts of drug 4.4. Influence of surfactant
– the EU directive recommends testing of concentrations up to
0.5 mg/ml. At typical SLN/NLC loading capacity of around 5–10% Surface properties of nanocarriers have decisive influence on
[5], the used concentrations are too low to cause any effect at all. their interactions with living cells and tissues. It is therefore obvi-
Another limiting factor in SLN/NLC testing in cell culture assays ous that surfactant type, amount and interaction with lipid core
is the highest SLN concentration in the cell culture media. A highly will have effect on SLN/NLC performance in cell cultures.
Table 2
Effect of blank SLN and NLC (i.e. without any encapsulated active ingredient) on cell viability expressed as % of viability reported by the authors of the study. This table includes the results that may be interpreted as not cytotoxic (cell
viability >70%). The column ‘‘drug’’ refers to the substance that the formulation was intended to encapsulate.

Type Lipid matrix Surfactant Active Size; PdI; ZP Cell line Assay Exposure Outcome (cell viability) Refs.
t (h) [] (mg/ml)a
SLN COM Epikuron + STC Ferulic acid LAN5 MTS 24 0.014– 95% to 0.056 mg/ml; 100% to [100]
0.056 other []
SLN COM/TM 0.5 wt% Polox 184/188/235/ n/a 186–280 nm; n.a.; HL-60 MTT 12 1  105– 100% up to 0.01 mg/ml [13]
335/407/T80/SDS n.a.; 1.0
SLN COM/TM 0.5 wt% Polox 184/188/235/ n/a 186–280 nm; n.a.; Granulocytes MTT 12 1  105– 100% up to 0.01 mg/ml [13]
335/407/T80/SDS n.a. 1.0
SLN CP/myristyl myristate (MM)/cetyl P188 n/a 185–197 nm; 3T3 BALB/c MTT 24 0.01–0.5 100% (CE up to 0.5 mg/ml); 85–65% [8]
esters (CE) 0.15–0.23; 30.5 (CP 0.01–0.5 mg/ml), 100–65%

S. Doktorovova et al. / European Journal of Pharmaceutics and Biopharmaceutics 87 (2014) 1–18


to 37.5 mV (MM 0.01–0.5 mg/ml)
SLN. SLN = 1%Apifil, P188 2.5% Alfa Lipoic Acid 103–121 nm; 0.18– NHF MTT 16 0.9 NE & 100% viability [46]
NLC, NLC = 8%Apifil + 1%Miglyol, 0.22, 27.8 to NLC; 0.1
NE NE = 9%Mygliol 36.8 mV SLN
SLN, NE SLN1 = CP, SLN2 = D114, P188 2.5% Gamma Oryzanol 200 nm; n.a.; n.a. NHF(NHDF?) MTT 48 1 100% viability [47]
SLN3 = WitepsolE85
SLN COM PC + T80 Mitoxantrone 108 nm; 0.219; MCF-7 MTT 72 100% viability [26]
26 mV
SLN TM PC, +STC PTC 224 nm; 0.278; n.a. MCF-7 MTT 72 100% viability [26]
SLN TP PC, +STC MTX 192 nm; 0.245; n.a. MCF-7 MTT 72 100% viability [26]
SLN+ Stearylalcohol/CTAB 2:1 or n/a 167 nm; 0.18; NCI/ADR- CellT- ? 0.1– 100% up to 1 lg CTAB, over 90% at [31]
Stearylalcohol/ceramide/CTAB 1:1:1 +39.3 mV or RES Glo 51.16 lg/ 51.16 lg CTAB/ml
75 nm; 0.21; ml CTAB
35.8 mV
SLN Stearylalcohol (ferrulate) T20 + ButOH + STC b-carotene/- 159 nm; 0.11; n.a RAT-1 MTT 24 1.32–13.52 100% up to 1.32 mg/ml, 81.5% at [48]
tocopherol 13.5 mg/ml, 58.1% 50 lM
Stearylalcohol ferrulate
SLN SA T80 + P188 Emodin 29 nm; n.a.; MCF-10A MTT n.a. n.a. 100% (all conc.) [88]
31 mV
SLN TM PC, Galactosylated DOPE DTC 120 nm; 0.2; 12.4 BEL7402 MTT 24 n.a. 100% (all conc.) [114]
to 13.2 mV
SLN CP T80 CPT n.a.; n.a.; n.a. BCECa AB 24 (0.25– 100% [115]
2.5 lM
CPT)
SLN TP T80 CPT n.a.; n.a.; n.a. BCECa AB 24 (0.25 lM 100% [115]
CPT)
SLN(?) Precirol 3 wt% T80 (50 wt%) PTC 114 nm; 0.117; MPMac MTT 2 0.075–7.5 100% [130]
20.1 mV
SLN(?) Precirol 3 wt% T80 (50 wt%) 114 nm; 0.117; MTX-B2 2 0.075–15 100% [130]
20.1 mV
SLN+ SA, stearylamine P188, Chol/PC/Chol + PC n.a. 272–305 nm; n.a.; HepG2 12 0.2 100% [131]
25–28 mV
SLN Precirol T80 Edelfosine 124 nm; 0.26; OCI-AML-2 Annexin- 24; 0.199 100% [132]
28.1 mV V 72
SLN COM P407/Poloxamine 908 n/a 200 nm, n.a.; n.a. Granulocytes MTT 12 1.0–25.0 100% (up to 1 mg/ml), 80–95% (up [12]
to 25 mg/ml)
SLN TP PTC 160 nm; 0.2; HL60 Trypan 48 2.5 No decrease in viability [49]
31 mV Blue
SLN SA DOX 80 nm; 0.2; HL60 Trypan 72 1 No decrease in viability [49]
35 mV Blue
SLN TP PTC 160 nm; 0.2; MCF-7 Trypan 48 2.5 No decrease in viability [49]
31 mV Blue
SLN SA DOX 80 nm; 0.2; MCF-7 Trypan 72 1 No decrease in viability [49]
35 mV Blue

(continued on next page)

7
8
Table 2 (continued)

Type Lipid matrix Surfactant Active Size; PdI; ZP Cell line Assay Exposure Outcome (cell viability) Refs.
t (h) [] (mg/ml)a
NLC COM (1.5 mg/ml) + Miglyol812 Epikuron (36 mg), STC (chemotherapeutic) 116 nm; 0.24; HuH6, HuH7 MTS 72 ? No cytotoxicity [116]
(0.3 mg/ml) (130.7 mg) 32.22 mV
NLC COM 1.50 mg/ml + Miglyol812 Epikuron (36 mg), STC (chemotherapeutic) 116 nm; 0.24; PC-3 MTS 72 ? No cytotoxicity [116]
0.3 mg/ml) (130.7 mg) 32.22 mV
SLN Hydrogenated Castor Oil 100 mg PVA 2 wt% Tilmicosin 189 nm; n.a.; COS-7 MTT 18 0. 5–4.0 No cytotoxicity [81]
7.8 mV
SLN Hydrogenated Castor Oil 100 mg PVA 2 wt% Tilmicosin 189 nm; n.a.; MDBK MTT 18 0. 5–4.0 No cytotoxicity (4 mg/ml [81]
7.8 mV decreased viability)
SLN(?)+ 5 mg/ml lipids (in wt%: 45 cholesteryl siRNA 117 nm; n.a.; MDAMB435 WST-8 24 0.003– 100% up to 0.048 mg/ml; 78 ± 1% [96]
ester, 3 TG, 14 DOPE, 10 (w/w) chol, 41.76 mV 0.072 at 0.072 mg/ml

S. Doktorovova et al. / European Journal of Pharmaceutics and Biopharmaceutics 87 (2014) 1–18


28 DC-chol)
SLN(?) E.wax, Brij78 5 mM (2 mg/ml lipids) PTC <100 nm; n.a.; n.a. U118 MTS 48, 0.023 No cytotoxicity [64]
72
SLN COM 0.3 wt%, StA 0.075 wt% 0.7 wt% ButOH n/a 100–200 nm; n.a.; CPH 54A Trypan 2 0.094 No toxicity up to 0.07 mg/ml [117]
(3.95 mg/ml) n.a. Blue
SLN COM 0.3 wt%, StA 0.075 wt% 0.7 wt% ButOH n/a 116 nm; 0.24; NR6-WA Trypan 2 0.094 No toxicity up to 0.07 mg/ml [117]
(3.95 mg/ml) 32.22 mV Blue
SLN COM 0.3 wt%, StA 0.075 wt% 0.7 wt% ButOH pDNA 116 nm; 0.24; CPH 54A Trypan 2 0.094 No toxicity up to 0.1 mg/ml [117]
(3.95 mg/ml) 32.22 mV Blue
SLN COM 0.3 wt%, StA 0.075 wt% 0.7 wt% ButOH pDNA 116 nm; 0.24; NR6-WA Trypan 2 0.094 No toxicity up to 0.1 mg/ml [117]
(3.95 mg/ml) 32.22 mV Blue
SLN COM Soy PC + P188 Epirubicin <450 nm; n.a.; n.a. A549 CV-A 24 ? No cytotoxicity [118]
SLN COM P188 + T80 CysA 227 nm; 0.214; n.a. Rabbit NR 2 2 99% [119]
cornea uptake
eptihelium
SLN+ SA, stearylamine P188, Chol/PC/Chol + PC n.a. 272–305 nm; n.a.; COS-1 PI 12 0.2 95% [131]
25–28 mV
SLN+ COM, Samine T80 Doxorubicin 127–151 nm; n.a.; A549 Trypan 48 0.002174 95% [133]
n.a. Blue
5
SLN TM PC PTC 129 nm; n.a.; OVCAR-3 MTT 24 14  10 – 90% highest [], 82.7 ± 7.4% at [20]
38.1 mV 14.0 100 lM Ptc
SLN(?) HSPC:DSPE:Chol:triolein = 1.5:1:1.2:1 P188 Curcumin/ 194 ± 2.89 nm; MCF-7 MTT 3, 6, ? >90% [21]
transferrin 0.15; 12.43 mV 12,
24,
48
SLN+ SA, stearylamine P188, Chol/PC/Chol + PC n.a. 272–305 nm; n.a.; Na1300 PI 12 0.2 90% [131]
25–28 mV
SLN Precirol T80 Edelfosine 124 nm; 0.26; HL60 Annexin- 72 0.199 90% [132]
28.1 mV V
SLN SA + PA PC Rifampicin 830 nm; 0.176; n.a. A549, MTT 24 0.03 90% [134]
NR8383,
pAEC, pAM
SLN Witepsol E85 P60 CPT n.a.; n.a.; n.a. BCECa AB 24 (0.25 lM >90% [115]
CPT)
SLN SA:isopropylmyristate = 3:1 T80 + TPGS + SoyPC + P188 Tripterine 93–321 nm; n.a.; Caco-2 MTT 36 ? >90% [120]
n.a.
SLN+ COM StA + T80 DOX 127–151 nm; A549 Trypan 48 ? >90% [69]
0.27–0.35; 27 mV Blue
SLN TS PC + T80 Ketoconazol 184–225 nm; 0.2; NIH-3T3 MTT 18 (1 mM >90% [121]
19 to 44 mV active)
SLN+ SA + StA P188 + PC + Chol n/a 336 nm; 0.38; HepG2 PI 12 1.0–3.0 >90% [122]
35 mV
SLN+ SA + StA P188 + Chol n/a 272 nm; 0.24; HepG2 PI 12 1.0–3.0 >90% [122]
28 mV
SLN COM/TM PC n/a HL-60 MTT 12 0.015–1.5 >90% to all []; (TM > COM viability) [13]
SLN COM P407/T80 n/a 268/214 nm; n.a.; HL-60 MTT 12 0.015–1.5 >90% Polox; 50% (T80 0.1 mg/ml); [13]
n.a. <40% 1.5 mg/ml
NLC COM + Miglyol 812 NTC Celecoxib 217 nm; 0.2; A549 CV-A 72 ? >90% [90]
27.38 mV
SLN CP/myristyl myristate (MM)/cetyl P188 n/a 185–197 nm; 3T3 BALB/c MTT 24 0.01–0.5 100% up to 0.5 mg/ml CE; 85–65% [8]
esters (CE) 0.15–0.23; 30.5 CP 0.01–0.5 mg/ml; 100–65% MM
to 37.5 mV 0.01–0.5 mg/ml
SLN SA PC + NTC DOX HT-29 72 (172 nM >90% v [65]
DOX)
NLC(?) Cacao butter + OA Pullulan-folate conjugate Pheo-A HeLa MTT 24 ? >90% NLC-Pheo A without access of [123]
light
SLN GMS PC + VitE + T80 DTC, ketoprofen, <100 nm; n.a.; n.a. bEnd.3 MTT 24 ? 97%, both SLN and SLN-folate [67]
folate targeted

S. Doktorovova et al. / European Journal of Pharmaceutics and Biopharmaceutics 87 (2014) 1–18


SLN GMS PC + VitE + T80 DTC, ketoprofen, <100 nm; n.a.; n.a. U373 24 ? 97%, both SLN and SLN-folate [67]
folate targeted
SLN SA PC + Myrj 59 BHR 75 nm; n.a.; 293T MTT 48 0.05–0.4 >95% viability [124]
2.5 mV
SLN SA PC + Myrj 59 BHR 75 nm; n.a.; HeLa MTT 48 0.05–0.4 >95% viability [124]
2.5 mV
SLN Witepsol (various) PC/T80 n/a 283–366 nm; n.a.; HaCaT MTT 18 0.15 >93.66% (all) [7]
26 to 32 mV
NLC(?) Chol oleate + Chol + trioleate PC Tan IIA 8 nm, n.a.; n.a.; RAW 264.7 MTT 3 0.03–0.15 95% (HDL-coated NLC), 76–88% [51]
viability
SLN SA PC + Myrj59 PPT d90 < 100 nm; n.a.; 293T MTT 24 0.1–0.4 >90% [50]
n.a.
SLN SA PC + Myrj59 PPT d90 < 100 nm; n.a.; HeLa MTT 24 0.1–0.4 >90% [50]
n.a.
SLN Softisan 100 PAA, PLL, Heparin Tenofovir VK2/E6E7 MTS 48 0.4–6.0 >80% (all conc.) [53]
SLN(?) Cacao butter/SA/CB + SA ButOH + CTAB + SDS + DSPE- DOX 200–250 nm; n.a.; U87MG XTT ? 0.8 88% (SA) – 95 (CB)% [70]
PEG-ca n.a.
SLN TM PC PTC 129 nm; n.a.; MCF-7 MTT 24 14  105– 80% (highest conc.) 70.7 ± 15% (at [20]
38.1 mV 14.0 equal 100 lM PTC)
SLN+ SA, stearylamine P188, Chol/PC/Chol + PC n/a 272–305 nm, n.a.; HepG2 PI 12 0.6 85% [131]
25–28 mV
SLN TP T20 + NTC Oryzalin 128 ± 5 nm; 0.12; THP1 MTT 24 ? >80% [91]
37 ± 4 mV
SLN TP PC + T20 + NTC Oryzalin 101 ± 4 nm; 0.22; THP1 MTT 24 ? >80% [91]
33 ± 2 mV
SLN TM/TL P188/P188 + NTC Ibuprofen 111 nm; n.a.; Caco-2 MTT 24 0.0–15.0 >80% [54]
+0.03 mV/121 nm;
n.a.; +0.02 mV
SLN+ SA + StA P188 + PC + Chol n/a 336 nm; 0.38; Na 1300 PI 12 1.0–3.0 >80% [122]
35 mV
SLN+ SA + StA P188 + PC n/a 305 nm; 0.24; HepG2 PI 12 1.0–3.0 >80% [122]
25 mV
SLN+ SA + StA P188 + PC + Chol n/a 336 nm; 0.38; COS-1 PI 12 1.0–2.0 >80% [122]
35 mV
SLN+ SA + StA P188 + Chol n/a 272 nm; 0.24; COS-1 PI 12 1.0–3.0 >80% [122]
28 mV
SLN CP/myristyl myristate (MM)/cetyl P188 n/a 185–197 nm; HaCaT MTT 24 0.01–0.5 80–70% (CP); 100–80% (MM); 100– [8]
esters (CE) 0.15–0.23; 30.5 85% (CE)
to 37.5 mV
SLN+ SA + CPC/SA + GMS + CPC P407 DOX n.a.; n.a.; 22– B16F10 MTT 24 1.32 80% [125]
40 mV
SLN CP, DA Octyldecanol, soy PC, P188 Etoposide 152 nm; n.a. K562 MTT 48 0.00625 80% [135]
>30 mV
SLN CP, SP Octyldecanol, soy PC, P188 Etoposide 171 nm; n.a.; K562 MTT 48 0.00625 80% [135]
25 mV

(continued on next page)

9
10
Table 2 (continued)

Type Lipid matrix Surfactant Active Size; PdI; ZP Cell line Assay Exposure Outcome (cell viability) Refs.
t (h) [] (mg/ml)a
SLN(?) Precirol 3 wt% T80 50 wt% 114 nm; 0.117; MPMac MTT 2 15 80% [135]
20.1 mV
SLN(?) HPO/TL/TP/docosanoic T80/T85/T20 n/a 61–103 nm, 0.39– BALB/c 3T3 MTT Over 80% viability [136]
0.47, 12.93 to
18.3 mV
SLN+ COM, Samine T80, Chol Retinoic acid 104–115 nm; n.a.; MCF-7, HCT- MTT 48 0.00625 80% [137]
n.a. 116 PBMC
NLC TP, OA T80 Simvastatin 149 nm; n.a.; HaCaT 72 0.3924 80% [138]
36 mV
SLN+ SA + StA + Chol P188 + PC + protamine n/a 336 nm; 0.38; COS-1 PI 12 0.1; 0.2; 80% (0.1 and 0.2 mg/ml), 70% at [52]

S. Doktorovova et al. / European Journal of Pharmaceutics and Biopharmaceutics 87 (2014) 1–18


35 mV 0.4 0.3 mg/ml
SLN SA T80 + P188 Emodin 29 nm; n.a.; MDA-MB- MTT 12, (up to >80% all times points and conc. [88]
31 mV 231 24, 30 lg
48 Emodin)
SLN SA T80 + P188 Emodin 29 nm; n.a.; MCF-7 MT 12, (up to >80% all times points and conc. [88]
31 mV 24, 30 lg
48 Emodin)
SLN+ GMS + CTAB/GM + LHNH PC n/a 50 nm; 0.1–0.23; A549 MTT 24, ? 80% CTAB-SLN; 90% LHNH SLN [36]
n.a. 48
SLN+ GMS + CTAB/GM + LHNH PC n/a 50 nm; 0.1–0.23; HeLa MTT 24, ? 80% CTAB-SLN; 90% LHNH SLN [36]
n.a. 48
SLN Witepsol (various) PC/T80 n/a 283–366 nm; n.a.; 3T3 MTT 18 Up to 0.15 >87.8% (all []) [7]
26 to 32 mV
SLN GMS + LHNH PC n/a 53 nm; n.a.; RAW 264.7 MTT 24 0.005–0.2 80–100% [33]
39.5 mV
SLN CP P407 n/a 200 nm; n.a.; n.a. Granulocytes MTT 12 25 80% [12]
SLN SA P188 Ibuprofen n.a.; 115; 0.32 mV Caco-2 MTT 24 0.0–15.0 >70% [54]
SLN+ SA + StA P188 + Chol n/a 272 nm; 0.24; Na 1300 PI 12 1.0–3.0 >70% [122]
28 mV
SLN+ SA + StA P188 + PC n/a 305 nm; 0.24; Na 1300 PI 12 1.0–3.0 >70% [122]
25 mV
SLN+ SA + StA P188 + PC n/a 305 nm; 0.24; COS-1 PI 12 1.0–2.0 >70% [122]
25 mV
SLN TM T80 CPT ? BCECa AB 24 (2.5 lM >70% [115]
CPT)
SLN CP, SA Octyldecanol, soy PC, P188 Etoposide 125 nm; n.a.; K562 MTT 48 0.006025 70% [135]
>40 mV
SLN CP, SP Octyldecanol, soy PC, P188 Etoposide 171 nm; n.a.; K562 MTT 48 0.0121 70% [135]
25 mV
SLN+ COM, Samine T80, Chol Retinoic acid 104–115 nm; n.a.; Jurkat MTT 48 0.007511 70% [137]
n.a.

Abbreviations (for this table): AB, alamar blue assay; BCEC, primary porcine brain capillary endothelial cells; BHR, Benzoic Acid; ButOH, butanol; CellT-Glo, Cell Titer Glo Assay (Luciferase& Luciferyl); CV-A, crystal violet assay;
E.wax, emulsifying wax; 2-Hydroxy-, 2-D-ribofuranosylhydrazide; MPMac, mouse peritoneal macrophages; MTT, MTT assay; NR, neutral red assay; PI, Propidium iodide exclusion assay; PPT, Podophyllotoxin; Tan IIA, Tanshinone
IIA; WST-8, WST-8 (Cell Counting Kit); [], concentration.
a
If not mentioned comes in mg/ml.
S. Doktorovova et al. / European Journal of Pharmaceutics and Biopharmaceutics 87 (2014) 1–18 11

Table 3
Effect of blank SLN and NLC formulation (i.e. without any encapsulated active ingredient) on cell viability expressed as % of viability reported by the authors of the study. This
table summarises the reported results that may be interpreted as cytotoxic (cell viability <70%) of SLN/NLC. The column ‘‘drug’’ refers to the substance that the formulation was
intended to encapsulate.

Type Lipid matrix Surfactant Active Size; PdI; ZP Cell Assay Exposure Outcome (cell viability) Refs.
line
t Conc.
(h) (mg/ml)a
SLN+ CP 3 wt%, DDAB T80:Span85 = 7:3 Plasmid n.a.; n.a.; n.a. HeLa AB 24 0.1–10.0 100% at 0.1 mg/ml 0.1–15% [126]
viability, above 0.4 mg/ml
SLN E.wax, Brij78 5 mM PTC <100 nm; n.a.; n.a. HCT- MTS 8– 0.023 68.6 ± 13.9% (24 h) [64]
(2 mg/ml lipids) 15 72 64.6 ± 10.5% (72 h)
SLN Witepsol E85 T60 CPT ? BCECa AB 24 (2.5 lM 50% [115]
CPT)
SLN CP, DA Octyldecanol, soy Etoposide 152 nm; n.a.; K562 MTT 48 0.0121 65% [135]
PC, P188 >30 mV
SLN+ SA, stearylamine P188, Chol/PC/ n/a 272–305 nm; n.a.; COS-1 PI 12 0.6 65% [131]
Chol + PC 25–28 mV
SLN CP 5 wt% T60/T80 2 wt% n/a 130–165 nm; <0.3; U251, MTT 24 0.05–5.0 <60% [32]
18 to 23 mV U373
SLN CP, DA Octyldecanol, soy Etoposide 152 nm; n.a.; K562 MTT 48 0.0241 60% [135]
PC, P188 >30 mV
SLN SA 0.33 wt% Lipoid/T80 n/a 283–366 nm; n.a.; 3T3 MTT 18 0.03 (1%) 40–64% [7]
26 to 32 mV
SLN SA 0.33 wt% Lipoid/T80 n/a 283–366 nm; n.a.; HaCaT MTT 18 0.03 (1%) 43–74% [7]
26 to 32 mV
SLN CP, SP Octyldecanol, soy Etoposide 171 nm; n.a.; 25 mV K562 MTT 48 0.0241 45% [135]
PC, P188
SLN CP, SA Octyldecanol, soy Etoposide 125 nm; n.a.; K562 MTT 48 0.0121 40% [135]
PC, P188 >40 mV
SLN(?) Precirol 3 wt% T80 50 wt% n/a 114 nm; 0.117; MPMac MTT 2 37.5 40% [130]
20.1 mV
SLN SA 0.33 wt% Lipoid/T80 n/a 283–366 nm; n.a.; J774 MTT 18 0.03 (1%) 8–18% [7]
26 to 32 mV
SLN(+) SA 0.33 wt%, StA n/a 284–381 nm; n.a.; J774 MTT 18 0.03 (1%) 1.45–2.47% [7]
0.066–0.13 wt% 18.0 to 24.0 mV
SLN COM 10 wt% PC 3 wt%, Risperidone n.a.; n.a.; n.a. Caco-2 MTT 24 40.6– 40–60% [28]
tyloxapol 1 wt% 550.0 lg/
cm2

Abbreviations for this table: BCEC, primary porcine brain capillary endothelial cells; MPMac, mouse peritoneal macrophages.
a
Concentration is given in mg/ml if no other unit is mentioned.

Fig. 3. Cell viability after 24 h exposure (in % viable cells) vs. concentration of SLN/
NLC in cell culture media. Data were extracted from that reported in the references Fig. 4. The effect of normal vs. cancer cell line on the outcome of cell viability
listed in Tables 1–3. From reports that list 3 and more different SLN/NLC studies after exposure to SLN. Data were extracted from references that used
concentration, the highest and lowest cell viabilities were used. ‘‘No decrease in normal (non-cancer) cell lines (open squares) and/or cancer cell lines (closed
cell viability’’ was plotted as 100% viability, ‘‘no toxicity’’ was plotted as 85% circles). Cell viability (in % viable cells) vs. concentration of SLN/NLC in cell culture
viability (as non-toxic could be between 70% and 100% viability). IC50 data are media reported in references listed in Tables 1–3 were used. From reports that list 3
plotted as 50% viability. and more different SLN/NLC concentration, the highest and lowest cell viabilities
were used. ‘‘No decrease in cell viability’’ was plotted as 100% viability, ‘‘no toxicity’’
was plotted as 85% viability (as non-toxic could be between 70% and 100% viability).
In a series of SLN composed of same lipid but different Polox- IC50 data are plotted as 50% viability.

amer (or Tween 80), slight differences were found among the for-
mulations, but none of them was toxic [13]. The surfactants used,
although non-toxic themselves (as illustrated by exposure to the The effect of surfactant influence is more pronounced with cat-
same cell line in the same report), could even improve their toler- ionic SLN/NLC, where the cationic surfactants/lipids alone show
ability when bound to SLN [30]. Binding of surfactant to SLN/NLC some toxicity towards living cells on the membrane level. Tabatt
surface decreases its availability to interact with cells and thus de- and collaborators [58] reported low viability of COS-1 cells after
creases its toxicity [13]. Schubert and Muller-Goymann however treatment SLN/NLC with positive surface charge; this was however
observed a contrary tendency in triglyceride-composed SLN, but still higher than when the same cells were treated with aqueous
in a smaller series of formulations [39]. solution of the same cationic surfactants. Single alkyl chain con-
12 S. Doktorovova et al. / European Journal of Pharmaceutics and Biopharmaceutics 87 (2014) 1–18

taining surfactants were found to be more toxic than double- efficiency of SLN and NLC for delivery of chemotherapeutic drugs
chained surfactants. Erni and collaborators reported that the viabil- was reviewed in detail elsewhere [74].
ity after solid lipid microparticles (SLM) treatment was similar to There might be various reasons for these observations: the che-
the treatments with aqueous cationic surfactant solutions [59], motherapeutic drugs, mostly highly lipophilic, are better soluble in
and, furthermore, noted that single-chain cationic surfactants lipid phase than in aqueous phase and therefore reach the cells in
coated particles had slightly smaller impact on the cells. These re- higher concentrations than from an aqueous solution, also the drug
sults might seem contradictory, but as different methods were can be protected by possible degradation by encapsulation [75].
used to obtain cell viability (Tabbat used LDH release which di- Furthermore, cellular uptake might be improved by encapsulation
rectly correlates to cell membrane damage, while Erni used ethi- into lipid nanoparticles of various types, not limited to SLN/NLC
dium bromide uptake assay) they are not to be automatically [57,76–78].
correlated. Although the paper of Erni and collaborators is entitled On the contrary to the majority of published results, Ying and
‘‘solid lipid microparticles’’, the composition of the nanoparticles is collaborators [37] reported that the IC50 of DOX-SLN was higher
relevant for SLN formulation and the resulting size (1.89–2.37 lm than that of DOX solution (after 48 h exposure). The authors justi-
for cationic SLM) was not much higher than the SLN size range, fied this finding by incomplete release of the drug from SLN. This is
therefore information from this study is still relevant also for eval- an important remark, once one reasons of encapsulation of drugs
uation of SLN/NLC with size <1 lm. into SLN/NLC is to achieve controlled release. Yet drug release is
mostly tested in different conditions than those found in cell cul-
ture (appropriate buffer solution or solvent mixture). It would be
4.5. SLN vs. NLC
interesting to see how well the in vitro release data correlate to ob-
served drug effects in cell culture.
Nanostructured lipid carriers contain, compared to SLN, addi-
The report by Carneiro and collaborators [79] shows that Treti-
tional oil component. Theoretically, the oil should be firmly incor-
noin (here tested for chemotherapeutic effect) loaded SLN had very
porated in the nanoparticle core and increase the stability of NLC
similar effect on cell viability of Jurkat, HCT-116, MCF-7 and nor-
by limiting the polymorphic phase changes during the storage
mal peripheral blood monocytes compared to blank SLN.
[2]. However, Jores and colleagues [60] found out that the oil
Also Wang and collaborators [80] tested a novel compound
may be attached to the solid lipid core surface. Although this
(2-hydroxy-, 2-D-ribofuranosylhydrazide, BHR) with possible anti-
‘‘spoon-like’’ appearance was also observed by Schwarz and col-
cancer effect in both normal and cancer-derived cell lines. In 293T
leagues [61], it is generally overseen in the literature. The implica-
cells, encapsulation into SLN could improve cell survival after
tions for tolerability in vitro include the availability for cell
treatment, compared to BHR solution. On the contrary, HeLa cells
interaction of another formulation component on the particle sur-
were more susceptible to BHR-loaded SLN treatment than BHR
face. The other question, yet without answer, is whether the oil,
solution. Treatment with blank SLN did not influence cell viability
adsorbed on the lipid core surface, stays on the surface in environ-
in neither of cell lines [80].
ment with higher ionic strength or if it suffers desorption as surfac-
tants may suffer [62].
4.7. SLN formulations for delivery of non-chemotherapeutic drugs
From the collected information, there is no apparent difference
between the toxicity of SLN and NLC. In Tables 1–3, NLC formula-
Evaluation of cytotoxicity of SLN and NLC formulations of other
tions can be found all around the tables, organised according to the
then cytotoxic drug is still not common, despite a large number of
toxicity. Only a few direct comparisons in SLN to NLC exist: 100%
existing SLN/NLC formulations. Again, there are also studies that
viability (in NHF cells) up to 0.1 mg/ml and 0.9 mg/ml for SLN vs.
state the drug-loaded formulation is not toxic, but do not disclose
NLC, respectively [46], and IC50 of 0.0264 mg/ml for SLN vs. IC50
the data of blank formulation – although in this case it is logical
of 0.0151 mg/ml for NLC in MDA-MB-231 cell line [63]. As these
that a non-toxic drug-loaded formulation is not toxic without the
only two available comparisons do not have concordant conclu-
drug, too. As the drugs approved for human use should be non-
sions, it is not possible to do a conclusion in favour of one of the
toxic per se, there are even few reports on SLN formulation only,
systems. NLC have improved characteristics in terms of drug load-
without testing the drug-loaded formulation – those are the fol-
ing and stability compared to SLN [2]; therefore, more data are to
lowing [53,81,82].
be expected in near future.
Considering the same fact, some of the reports of non-chemo-
therapeutic drugs which are non-toxic in vitro, are raising some
4.6. SLN formulations for delivery of chemotherapeutic drugs concern. Alpha-lipoic acid, on its own non-toxic to NHF cells over
the concentration range used (0.01–10.0 lM), caused decrease in
Almost all available reports about SLN/NLC loaded with chemo- cell viability by 30% when loaded into NLC and by 20% when loaded
therapeutic drugs state that despite blank formulations were not into Apifil-composed SLN. The blank formulations were both very
toxic to the cell line in use, upon loading with the drug the toxicity well tolerated (no decrease in cell viability up to 0.1 mg/ml for
exceeded the toxicity of drug solution alone. There are examples SLN and 0.9 mg/ml for NLC) [46]. Similar effect was observed in
for paclitaxel [20,49,64–66], docetaxel [55,67,68], doxorubicin the same cell line by the same research group when examining a
[49,69,70], mitoxantrone [71] and others. Surface modified or gamma-oryzanol SLN formulation. The drug, by itself non-toxic,
targeted formulations showed even greater toxicity than non- and non-toxic when encapsulated in CP-composed SLN, while
targeted formulation [21,72]. Increased susceptibility to chemo- when encapsulated into TM-composed SLN, the cell viability
therapeutic drugs administered in SLN/NLC was reported also in dropped to 60% [47]. A time-dependent and dose dependent toxic-
resistant cell lines [64,66]. Blank NLC were shown to be non-toxic ity was observed for beta-carotene encapsulated in otherwise very
to the cell line in question (MCF-7 vs. MCF-7/MX resistant to mito- well tolerated (80% cell viability at 13.5 mg/ml) formulation in
xanthrone). Viability below 70% was reported for paclitaxel resis- RAT-1 cells [48].
tant HCT-15 cell line [64]. A study by Baek and Cho suggests that On the other hand, Souza and collaborators [83] reported im-
encapsulation of paclitaxel in SLN caused similar improvement in proved cell viability of HepG2 cells treated with praziquantel-
drug effect as pre-treatment with a p-glycoprotein inhibitor loaded SLN compared to those treated with praziquantel solution
(verapamil), and that combination of verapamil pre-treatment (in ethanol). The authors raised questions of the observed toxicity
and encapsulation could further improve the outcome [73]. The to be caused by the solvent used for preparation of drug solution,
S. Doktorovova et al. / European Journal of Pharmaceutics and Biopharmaceutics 87 (2014) 1–18 13

despite it being applied in minimal amounts [75,83]. Similar obser- processes (endocytosis) might be involved in SLN uptake [29]. By
vation was reported by Teskac and Kristl with resveratrol-loaded pre-incubation with specific uptake mechanism inhibitors, two
SLN and tripterin-loaded NLC [84]. Resveratrol-loaded SLN in this independent research group could specify that clathrin-mediated
case caused even higher rate of keratinocytes cell growth than that endocytosis might be the preferential uptake mechanism, while
observed in untreated control and drug-solution treated control caveolae-mediated endocytosis seems to play only minor role or
[85]. not involved at all [32,71]. Martins and collaborators also found
Li and colleagues [86] observed higher cytotoxicity of their buf- out that macropinocytosis may be involved to smaller extent in
adienolides-loaded NLC after first 48 h of treatment than free some cell types.
drugs; at 72 h and 96 h more damage to HGC-27 cells was caused
by free drug than NLC. The authors observed that this behaviour
6. Membrane integrity
correlated with the release pattern of the drug, showing a burst re-
lease during the first 8 h and then showed sustained release at
LDH release assay was used in several reports on SLN/NLC
steady rate, releasing 85% of encapsulated drug [86].
safety. Many of the authors used it as a measure of cell viability,
Other authors reported absence of toxicity of neither blank nor
most of them alongside with a formazan-dye based assay. These
drug loaded SLN [53,54,87–91]. Wolf and collaborators confirmed
authors concluded that LDH assay was less sensitive to potential
excellent tolerability of SLN in keratinocytes, skin fibroblasts and
effects on cell than WST-1 [95] or MTT assay [10,29], from which
in a reconstructed human epidermis model (Episkin), showing that
result higher IC50 values.
the system is harmless to skin cells and can be used for therapy of
Alukda and colleagues reported no increase in LDH release com-
skin diseases and wounds [92].
pared to control in the range of 0.4–6.0 mg/ml SLN in VK2/E6E7
These contradictory results imply the importance of toxicity
cells after 48 h exposure, while MTS assay revealed a decrease in
testing for each particular formulation. While the effect of in-
cell viability (80% viable cells) [53]. Even greater differences where
creased toxicity upon encapsulation in SLN/NLC can again be ex-
quite surprisingly reported by Olbrich and collaborators of cationic
plained by the fact that the drugs are better solubilised and thus
SLN: while IC50 for Esterqaut-containing SLN was 0.6–0.7 mg/ml by
available in higher concentration in the system, on the other hand,
WST-1, LDH assay indicated 3.0–5.0 mg/ml as IC50 [95].
the effect of reduced cytotoxicity may be explained by incomplete
This difference in sensitivity can also be interpreted as higher
release of the drug from the particles.
susceptibility of mitochondrial enzymes (responsible for formazan
dye metabolism) to SLN/NLC presence than the cellular membrane,
4.8. Comparison to other types of colloidal carriers or by decrease in mitochondrial metabolism and other intracellular
pathways occurrence prior cell membrane rupture.
As evidenced already, in the very first years of development of Montenegro and colleagues [35] used LDH-assay to examine the
SLN and later NLC, the biocompatibility of these systems is remark- ability of idebenone loaded SLN to protect the cells (primary astro-
ably better as that of comparable colloidal systems prepared from cytes) from oxidative damage induced by 2,20 -azobis-(2-amidino-
polymers. First of all, the lipid material used is biodegradable and propane) dihydrochloride (AAPH). By increasing the dose, the
thus does not accumulate in the tissue/organism. Secondly, the LDH release values could return to values similar to AAPH
typical average size of SLN/NLC between 100 nm and 1 lm, by untreated group [35].
some not even considered as ‘‘nanoscale’’, was until recently con-
sidered too large for cells to internalise [93]. Recent insights how-
7. ROS generation
ever prove that even non-phagocytic cells are able to effectively
internalise objects as large as 2–3 lm [94].
The presence of nanoparticles and their interaction with biolog-
Direct comparison of SLN to other colloidal drug (or gene) deliv-
ical membranes can lead to such disturbances in cell metabolism
ery systems in terms of safety favours lipid nanoparticles. While
that lead to increased reactive oxygen species (ROS) production.
0.35 mg/ml of polycyanoacrylate nanoparticles was a dose 100%
Many reports on increased oxidative stress in cells in culture after
lethal for HL-60 macrophages, 1.5 mg/ml SLN could be well toler-
exposure to nanomaterials exist (for example, see [44,97]). In case
ated (90% cell viability). The lethal dose of PMCA-NP towards HL-
of SLN/NLC, this issue has not been addressed yet in a manner
60 in the same study was even smaller – 0.05 mg/ml [13]. IC50 of
allowing for a general conclusion. As increased ROS may not neces-
poly-L-lysine (22 kDa) and poly-ethyleneimine (PEI, 25 kDa) in
sarily lead to cell death, it may occur also in the cells and can be
COS-1 cells was 10 lg/ml and 20 lg/ml, while IC50 of cationic
induced by formulations that have been successfully tested by cell
SLN was 0.6–0.7 mg/ml, respectively, depending on the solid lipid
viability methods.
used [95]. PEI was also compared to SLN in MDA-MD-435 cells,
From the few existing report addressing the oxidative stress is-
the outcome being IC50 of 0.009 mg/ml for PEI and no toxicity
sue, most of them used the DCFH-DA method. DCFH-DA (20 ,70 -
caused by SLN at 0.048 mg/ml [96].
Dichlorofluorescein diacetate) is a highly permeable substance that
enters cells easily. In the presence of oxidants, it is metabolised
5. Uptake by cells into fluorescent product DCFH. However, DCFH-DA can react with
a variety of radicals present in the cells, and it is prone to interfer-
Evaluation of all of the effects on living cells in culture has the ence [98]. Other methods for detection of free radicals and their ef-
pre-requisite of uptake of the particles by the cells, or interaction of fects were reviewed elsewhere [99]. Mulik and collaborators
SLN/NLC components with the cells. As documented by many re- reported that the increase in ROS content after SLN exposure was
ports, SLN and NLC are promptly internalised by a wide varieties only negligible compared to untreated MCF-7 cells [21]. Picone
of cell lines, including cancer cell lines [32,57,76], non-cancer cell and colleagues also reported ROS generation levels near to control
lines [32,85], and both phagocytic and non-phagocytic cells. [100]. In addition, ferulic acid loaded SLN could protect the COS-1
Attempts to clarify the uptake mechanism have been published cells better than ferulic acid solution. This effect was found dose-
recently. Coumarin-6 marked SLN could be well internalised into dependent [101]. Idebenone-loaded SLN also showed strong pro-
A30 cells at 37 °C. The uptake was decreased but not completely tective effect against AAPH induced ROS generation [35]. Gökce
stopped at 4 °C incubation, which suggests that energy-dependent and colleagues reported that ROS level in skin fibroblasts was
14 S. Doktorovova et al. / European Journal of Pharmaceutics and Biopharmaceutics 87 (2014) 1–18

lower after treatment with coenzyme Q10-loaded SLN than after 10. Blood compatibility (red blood cells)
treatment with Q10 alone [102].
In contradiction to other reports [21,100], Silva and collabora- Important information about safety of formulations intended
tors [15] reported that ROS generation associated to SLN exposure for intravenous administration is the compatibility with red blood
may occur. The cells, when pre-incubated with N-acetylcysteine cells (RBCs). There are some reports on RBC compatibility testing,
(an antioxidant) and/or catalase had significantly higher viability and the results are all very encouraging. Studies by Schubert and
after SLN exposure [15], meaning that these antioxidants could Muller-Goymann [39] and Li and collaborators [86] reported a very
protect the cells from SLN induced loss of viability. low % of released oxy-haemoglobin – RBCs exposed SLN composed
Very interesting in vivo results were reported by Kakkar and of Softisan 100 and varying content of phospholipids up to 50% of
Kaur [103] in a murine model of Alzheimer disease. After AlCl3 lipid matrix, stabilised by Solutol HS15 presented only (1.8 ± 1.6)%
damage induction of damage, the levels of oxidised lipids were in- release, which corresponded to about 0.4% of positive control (dis-
creased, reduced glutathione (GSH) pool decreased and activity of tilled water) [39]. Treatment with NLC composed of GMS, OA and
superoxide dismutase and catalase (which are the enzymes in- Miglyol 812, stabilised with a combination of P188, phosphatidyl-
volved in cellular protection against ROS) increased. Treatment choline (PC) and even SDS, lead to less than 5% haemolysis at
with curcumin-loaded SLN could reverse these changes. Interest- 1.2 mg/ml SLN, depending on the final SLN amount [86].
ingly, a reverse to values close to normal was also reported after Koziara and collaborators showed that, similarly to cell viability
administration of blank SLN only [103]. in vitro, the effect of surfactant alone is reduced when used in SLN.
Time dependent and concentration effect was observed after treat-
ment with Brij 78 in solution in the same amount as used in SLN.
8. Genotoxicity While haemolysis ratio increased with time and concentration up
to 80% (6 h, highest concentration), haemolysis after 1 mg/ml SLN
One of the sub-lethal effects of nanoparticles, addressed treatment was still below 10% [108]. Further reports exist on actu-
throughout in the research of inorganic nanoparticles is the dam- ally lower haemolysis of drug-loaded SLN/NLC than free drugs –
age to DNA. DNA damage can include oxidation of the bases, strand DOX caused higher extent of haemolysis than DOX-SLN, even smal-
breaks or disturbances in DNA repair. A fixation of an error in DNA ler impact was observed after mannosylated DOX-SLN treatment
sequence is the pre-requisite for mutation development. Our group [109]. Same tendency was reported for SLN loaded with metoth-
tested a small set of cationic SLN in two cell lines by comet assay rexate [110]. It is interesting that both SLN formulations in the last
(for method details see [104,105]. Although we found a small insig- two mentioned reposts had positive surface charge, and their sur-
nificant increase in comet tail intensity (which represent the DNA faced variant (mannosylated, albumin-covered) was also success-
strand breaks or the sites with defect or missing bases converted fully tested.
into strand breaks), the overall result allowed to conclude that Taken together, there seems to be no reason for concerns about
these formulations were not genotoxic. negative effects on RBSc.
The material used for SLN/NLC production gives the pre-requi-
site of a well tolerated carrier also in the perspective of genotoxi-
city, however it would be interesting to confirm the absence of
genotoxicity by more studies. As DNA damage is often linked to in- 11. Safety in vivo?
creased oxidative damage [106], and SLN may have some effect on
ROS generation (increase, as reported by [15]), and many reports Despite large number of published in vivo studies, the large
showed SLN reaching perinuclear region of the cells, the genotox- majority focuses on SLN efficiency rather than their safety. Among
icity studies might have interesting outcomes. the few in vivo toxicological studies, there are even fewer that al-
low for comparison of the dose used in in vivo experiments with
the observed toxicity in vitro in cell culture. Therefore, from the
9. Phagocytic cells, inflammation response available data, it is not possible to establish a link between a ‘‘safe’’
SLN/NLC concentration for in vitro and in vivo.
SLN uptake by macrophages which is not desired as it leads to Tolerability of SLN by airways and lung tissue were examined in
rapid elimination from the blood circulation was tested in three re- mice and rats. Nassimi and collaborators [9] tested tolerability
ports. SLN stabilised by Poloxamer 188 (polyethylene–polypropyl- both in vitro and in vivo. In vitro assays revealed IC50 in A549 cells
ene glycol) or Myrj (polyethylene glycol monostearate) are of 2 mg/ml (by NR uptake assay) or 3 mg/ml (by MTT assay). Doses
expected to have ‘‘stealth’’ characteristics and avoid phagocytosis between 1 and 200 lg, of lung-deposited solids, were tested in fe-
for a prolonged time. In in vitro and in vivo studies, murine macro- male BALB/c mice. SLN did not trigger inflammation; neither LDH
phages could internalise such SLN to a smaller extent; the peak activity in bronchoalveolar lavage was increased. Microscopic
absorption occurred at 120 min in both studies [51,107]. Wang appearance of the lung parenchyma was found normal; although
and collaborators reported that increasing Myrj concentration in some minor aggregates of lipid-filled macrophages were found [9].
formulation and/or polyoxyethylene chain length of Myrj could re- Although cytotoxicity of blank NLC was not tested in cell lines
duce the uptake [107]. Zhang and colleagues [51] found out that (Bufadienolides-loaded NLC were cytotoxic to HGC-27 and U87-
phagocytosis of SLN stabilised with Poloxamer 188 occurred to MG cells, presenting similar IC50 values as Bufadienolides solution
the same extent independently of the presence of serum in the cul- in DMSO), a LD50 in mice was 1.15 higher than LD50 of drug solu-
ture medium, while non-stealth SLN were internalised to higher tion (2.31 mg/kg and 2.95 mg/kg for drug solution and NLC, respec-
extend in serum presence in the culture medium. Phagocytic up- tively). Strong presentation of toxic effects in mice administered
take of SLN by normal human granulocytes measured by luminal with drug solution was observed at 1.32 mg/kg drug, while no
assay could be highly decreased compared to luminal control. Fur- marked toxic effects were observed after receiving bufadieno-
thermore, Poloxamine 908 stabilised-SLN was shown more resis- lides-NLC [86]. Fangueiro and colleagues [112] tested the effect
tant against the uptake than Poloxamer 407 covered SLN [12]. of a SLN formulation on Drosophila melanogaster survival and
This is interesting information and raises a question, why polox- reproduction. Although small reduction in number of progeny flies
amine 908 is only rarely used in SLN formulations intended for was detected, this was similar to the effect of bulk lipid (Softisan
intravenous administration? 145) admixed in the standard Drosphila medium.
S. Doktorovova et al. / European Journal of Pharmaceutics and Biopharmaceutics 87 (2014) 1–18 15

A good correlation between in vitro and in vivo results was re- grant to SD) and PEst-OE/EQB/LA0023/2011 is acknowledged.
ported by Olbrich and collaborators [113] with SLN used as vaccine AMS was supported by European Union Funds (FEDER/COMPETE)
adjuvants in chicken. A cationic SLN formulation (solid paraf- and by national funds (FCT) under the project FCOMP-01-0124-
fin + esterquat1 (EQ), stabilised by T80 + Span85 mixture) which FEDER-022696 and by a research project grant (PEst-C/AGR/
was toxic towards RAW 246.7 macrophages in vitro (1 mg/ml), also UI4033/2011). The authors also wish to acknowledge FCT, under
caused significant damage in tissue at injection site (dose 7.2 mg/ the reference PTDC/SAU-FAR/113100/2009.
ml). This damage was still lower when compared to standard
vaccine adjuvant. This formulation caused decrease in viability in
human foreskin fibroblast, but by no more than 20%. On the con- References
trary, a negatively charged formulation without EQ, non-toxic
[1] L. Battaglia, M. Gallarate, Lipid nanoparticles: state of the art, new preparation
in vitro, was also well tolerated in chicken [113]. methods and challenges in drug delivery, Exp. Opin. Drug Deliv. 9 (2012)
497–508.
[2] R. Muller, R. Shegokar, C. Keck, 20 Years of lipid nanoparticles (SLN and NLC):
12. Conclusions and further perspectives present state of development and industrial applications, Curr. Drug Discov.
Technol. 8 (2011) 207–227.
[3] R.H. Müller, M. Radtke, S.A. Wissing, Nanostructured lipid matrices for
As shown in this review, great amount of work has been done improved microencapsulation of drugs, Int. J. Pharm. 242 (2002) 121–128.
and there is large amount of in vitro data available. The basic con- [4] J. Pardeike, A. Hommoss, R.H. Muller, Lipid nanoparticles (SLN, NLC) in
cosmetic and pharmaceutical dermal products, Int. J. Pharm. 366 (2009) 170–
cept of SLN and NLC as a well tolerated carrier seems to be con-
184.
firmed and well documented. When it comes to more detailed [5] W. Mehnert, K. Mader, Solid lipid nanoparticles: production, characterization
questions, the currently available data still do not offer straightfor- and applications, Adv. Drug Deliv. Rev. 47 (2001) 165–196.
ward answers. Some researchers questioned if cancer cell lines are [6] J.S. Lucks, R.H. Müller, B. König, Solid lipid nanoparticles (SLN) – an alternative
parenteral drug carrier system, Eur. J. Pharm. Biopharm. 38 (1992) 33S.
more susceptible to SLN/NLC than ‘‘normal’’ cell lines, but with cur- [7] W. Weyenberg, P. Filev, D. Van den Plas, J. Vandervoort, K. De Smet, P. Sollie,
rently available data one should be very careful to draw a conclu- A. Ludwig, Cytotoxicity of submicron emulsions and solid lipid nanoparticles
sion like this. ‘‘Normal’’ cell lines, despite being derived from for dermal application, Int. J. Pharm. 337 (2007) 291–298.
[8] D.M. Ridolfi, P.D. Marcato, D. Machado, R.A. Silva, G.Z. Justo, N. Duran, In vitro
healthy tissue, still represent a set of dividing cells that does not cytotoxicity assays of solid lipid nanoparticles in epithelial and dermal cells, J.
normally occur with mature differentiated cells. To the best of Phys: Conf. Ser. 304 (2012) 012032.
our knowledge, no study on ‘‘normal’’ cells exists up to date and [9] M. Nassimi, C. Schleh, H.D. Lauenstein, R. Hussein, H.G. Hoymann, W. Koch, G.
Pohlmann, N. Krug, K. Sewald, S. Rittinghausen, A. Braun, C. Müller-Goymann,
it is a questionable if it would to be feasible with nanocarriers that A toxicological evaluation of inhaled solid lipid nanoparticles used as a
are not expected to cause any damage. Different situation is with potential drug delivery system for the lung, Eur. J. Pharm. Biopharm. 75
leukocytes, where normal macrophages and granulocytes from hu- (2010) 107–116.
[10] M. Nassimi, C. Schleh, H.-D. Lauenstein, R. Hussein, K. Lübbers, G. Pohlmann,
man healthy volunteers appear much more susceptible to SLN/NLC S. Switalla, K. Sewald, M. Müller, N. Krug, C.C. Müller-Goymann, A. Braun, Low
than leukaemia-derived HL-60 or virus containing RAW 264.7 cytotoxicity of solid lipid nanoparticles in in vitro and ex vivo lung models,
macrophages [12,13]. Inhal. Toxicol. 21 (2009) 104–109.
[11] C. Olbrich, N. Scholer, K. Tabatt, O. Kayser, R.H. Muller, Cytotoxicity studies of
We divided the SLN/NLC in vitro data also by the potential active
Dynasan 114 solid lipid nanoparticles (SLN) on RAW 264.7 macrophages-
substance they might carry by purpose. Except from one report, all impact of phagocytosis on viability and cytokine production, J. Pharm.
studies on chemotherapeutic drug loaded SLN/NLC report an in- Pharmacol. 56 (2004) 883–891.
crease in toxicity towards the target cancer cells. However, the [12] R.H. Muller, S. Maassen, H. Weyhers, W. Mehnert, Phagocytic uptake and
cytotoxicity of solid lipid nanoparticles (SLN) sterically stabilized with
other group of formulations for non-chemotherapeutic drugs refers poloxamine 908 and poloxamer 407, J. Drug Target. 4 (1996) 161–170.
to both increase and decrease in toxicity. [13] R.H. Muller, D. Ruhl, S. Runge, K. Schulze-Forster, W. Mehnert, Cytotoxicity of
Taken altogether, prediction of action of a drug encapsulated in solid lipid nanoparticles as a function of the lipid matrix and the surfactant,
Pharm. Res. 14 (1997) 458–462.
SLN/NLC can still be tricky. We think that the contradictory results [14] S. Petersen, F. Steiniger, D. Fischer, A. Fahr, H. Bunjes, The physical state of
are also due to some bias of the studies of chemotherapeutic drugs. lipid nanoparticles influences their effect on in vitro cell viability, Eur. J.
Furthermore, all of these data are only a small hint on how SLN/ Pharm. Biopharm. 79 (2011) 150–161.
[15] A.H. Silva, F.B. Filippin-Monteiro, B. Mattei, B.G. Zanetti-Ramos, T.B.
NLC will behave in vivo. We still have almost no information on Creczynski-Pasa, In vitro biocompatibility of solid lipid nanoparticles, Sci.
side effects at site of administration (except from dermal products Total Environ. 432 (2012) 382–388.
which were already tested on human volunteers), and the systemic [16] International Organization for Standardization, ISO 10993-5:2009 Biological
Evaluation of Medical Devices Part 5: Tests for in Vitro Cytotoxicity, 2009.
toxicity on the organism as whole. More important questions [17] N. Lewinski, V. Colvin, R. Drezek, Cytotoxicity of nanoparticles, Small 4 (2008)
would arise when considering per oral route of administration, as 26–49.
– although biodegradable – an increased uptake of lipids will have [18] T. Mosmann, Rapid colorimetric assay for cellular growth and survival:
application to proliferation and cytotoxicity assays, J. Immunol. Methods 65
an effect on the organism in question.
(1983) 55–63.
To summarise all, a large amount of in vitro data about SLN and [19] S. Gattefosse, Precirol ATO 5 Technical data sheet, in: S. Gattefosse (Ed.),
NLC is already available, but the overall question if SLN/NLC are France, 2009.
really so well tolerated at any circumstances is still not answered. [20] M.-K. Lee, S.-J. Lim, C.-K. Kim, Preparation, characterization and in vitro
cytotoxicity of paclitaxel-loaded sterically stabilized solid lipid nanoparticles,
We would like to draw the attention to the fact that there are many Biomaterials 28 (2007) 2137–2146.
contradictory results, and also many results which might suffer for [21] R.S. Mulik, J. Monkkonen, R.O. Juvonen, K.R. Mahadik, A.R. Paradkar,
some influence of the actual purpose of the SLN/NLC formulation. Transferrin mediated solid lipid nanoparticles containing curcumin:
enhanced in vitro anticancer activity by induction of apoptosis, Int. J.
Animal studies with SLN/NLC are reaching almost the same num- Pharm. 398 (2010) 190–203.
ber as in vitro studies reviewed here. Once the authors also focus [22] G. Repetto, A. del Peso, J.L. Zurita, Neutral red uptake assay for the estimation
on screening for possible side effects or their absence apart from of cell viability/cytotoxicity, Nat. Protoc. 3 (2008) 1125–1131.
[23] W. Strober, Trypan blue exclusion test of cell viability, in: John E. Coligan,
efficiency, bio-distribution and pharmacokinetics, only then it et al. (Eds.), Current Protocols in Immunology, Appendix 3, Appendix 3B,
would be able to confirm that SLN and NLC are safe nanocarriers. 2001.
[24] G. Fotakis, J.A. Timbrell, In vitro cytotoxicity assays: comparison of LDH,
neutral red, MTT and protein assay in hepatoma cell lines following exposure
Acknowledgments to cadmium chloride, Toxicol. Lett. 160 (2006) 171–177.
[25] Q.H. Ma, Y.W. Wang, X.F. Lin, D. Luo, N. Gu, Preparation, characterization and
photoprotection of tocopherol loaded nanostructured lipid carriers, in: IEEE/
Financial support from Portuguese Science and Technology ICME International Conference on Complex Medical Engineering, 2007, CME
Foundation (FCT) under reference SFRH/BD/60552/2009 (Doctoral 2007, 2007, pp. 203–208.
16 S. Doktorovova et al. / European Journal of Pharmaceutics and Biopharmaceutics 87 (2014) 1–18

[26] Y. Zhuang, B. Xu, F. Huang, J. Wu, S. Chen, Solid lipid nanoparticles of podophyllotoxin-loaded solid lipid nanoparticles, Nanotechnology 20
anticancer drugs against MCF-7 cell line and a murine breast cancer model, (2009) 055702.
Pharmazie 67 (2012) 925–929. [51] W. Zhang, J. Liu, S. Li, M. Chen, H. Liu, Preparation and evaluation of stealth
[27] C. Freitas, R.H. Muller, Stability determination of solid lipid nanoparticles Tashinone IIA-loaded solid lipid nanoparticles: influence of Poloxamer 188
(SLN) in aqueous dispersion after addition of electrolyte, J. Microencapsul. 16 coating on phagocytic uptake, J. Microencapsul. 25 (2008) 203–209.
(1999) 59–71. [52] E. Vighi, E. Leo, M. Montanari, A. Mucci, M. Hanuskova, V. Iannuccelli,
[28] A.C. Silva, A. Kumar, W. Wild, D. Ferreira, D. Santos, B. Forbes, Long-term Structural investigation and intracellular trafficking of a novel
stability, biocompatibility and oral delivery potential of risperidone-loaded multicomposite cationic solid lipid nanoparticle platform as a pDNA carrier,
solid lipid nanoparticles, Int. J. Pharm. 436 (2012) 798–805. Ther. Deliv. 2 (2011) 1419–1435.
[29] I. Rivolta, A. Panariti, B. Lettiero, S. Sesana, P. Gasco, M. Gasco, M. Masserini, G. [53] D. Alukda, T. Sturgis, B. Youan, Formulation of tenofovir-loaded
Miserocchi, Cellular uptake of coumarin-6 as a model drug loaded in solid functionalized solid lipid nanoparticles intended for HIV prevention, J.
lipid nanoparticles, J. Physiol. Pharmacol. 62 (2011) 45–53. Pharm. Sci. 100 (2011) 3345–3356.
[30] C.W. How, R. Abdullah, R. Abbasalipourkabir, Characterization and [54] S.G. Potta, S. Minemi, R.K. Nukala, C. Peinado, D.A. Lamprou, A. Urquhart, D.
cytotoxicity of nanostructured lipid carriers formulated with olive oil, Douroumis, Preparation and characterization of ibuprofen solid lipid
hydrogenated palm oil and Polysorbate 80, IEEE Trans. Nanobiosci. 12 nanoparticles with enhanced solubility, J. Microencapsul. 28 (2010) 74–81.
(2013) 72–78. [55] D. Liu, Z. Liu, L. Wang, C. Zhang, N. Zhang, Nanostructured lipid carriers as
[31] A. Siddiqui, G.A. Patwardhan, Y.-Y. Liu, S. Nazzal, Mixed backbone antisense novel carrier for parenteral delivery of docetaxel, Colloids Surf., B:
glucosylceramide synthase oligonucleotide (MBO-asGCS) loaded solid lipid Biointerfaces 85 (2011) 262–269.
nanoparticles: in vitro characterization and reversal of multidrug resistance [56] N. Schöler, H. Hahn, R.H. Müller, O. Liesenfeld, Effect of lipid matrix and size
in NCI/ADR-RES cells, Int. J. Pharm. 400 (2010) 251–259. of solid lipid nanoparticles (SLN) on the viability and cytokine production of
[32] S. Martins, S. Costa-Lima, T. Carneiro, A. Cordeiro-da-Silva, E.B. Souto, D.C. macrophages, Int. J. Pharm. 231 (2002) 167–176.
Ferreira, Solid lipid nanoparticles as intracellular drug transporters: an [57] H. Yuan, J. Miao, Y.Z. Du, J. You, F.Q. Hu, S. Zeng, Cellular uptake of solid lipid
investigation of the uptake mechanism and pathway, Int. J. Pharm. 430 nanoparticles and cytotoxicity of encapsulated paclitaxel in A549 cancer
(2012) 216–227. cells, Int. J. Pharm. 348 (2008) 137–145.
[33] W. Yu, C. Liu, Y. Liu, N. Zhang, W. Xu, Mannan-modified solid lipid [58] K. Tabatt, M. Sameti, C. Olbrich, R.H. Müller, C.-M. Lehr, Effect of cationic lipid
nanoparticles for targeted gene delivery to Alveolar macrophages, and matrix lipid composition on solid lipid nanoparticle-mediated gene
Pharmaceut. Res. 27 (2010) 1584–1596. transfer, Eur. J. Pharm. Biopharm. 57 (2004) 155–162.
[34] J.A.R.d. Araújo, in: Triamcinolone Acetonide for Ocular Administration: [59] C. Erni, C. Suard, S. Freitas, D. Dreher, H.P. Merkle, E. Walter, Evaluation of
Innovations in Drug Delivery and Therapeutics, Departament de cationic solid lipid microparticles as synthetic carriers for the targeted
Físicoquímica, Universitat de Barcelona, Barcelona, Spain, 2011. delivery of macromolecules to phagocytic antigen-presenting cells,
[35] L. Montenegro, A. Campisi, M.G. Sarpietro, C. Carbone, R. Acquaviva, G. Raciti, Biomaterials 23 (2002) 4667–4676.
G. Puglisi, In vitro evaluation of idebenone-loaded solid lipid nanoparticles [60] K. Jores, W. Mehnert, M. Drechsler, H. Bunjes, C. Johann, K. Mäder,
for drug delivery to the brain, Drug Dev. Ind. Pharm. 37 (2011) 737–746. Investigations on the structure of solid lipid nanoparticles (SLN) and oil-
[36] W. Yu, C. Liu, J. Ye, W. Zou, N. Zhang, X. Wenfang, Novel cationic SLN loaded solid lipid nanoparticles by photon correlation spectroscopy, field-
containing a synthesized single-tailed lipid as a modifier for gene delivery, flow fractionation and transmission electron microscopy, J. Control. Release
Nanotechnology 20 (2009) 215102. 95 (2004) 217–227.
[37] X.-Y. Ying, D. Cui, L. Yu, Y.-Z. Du, Solid lipid nanoparticles modified with [61] J.C. Schwarz, A. Weixelbaum, E. Pagitsch, M. Low, G.P. Resch, C. Valenta,
chitosan oligosaccharides for the controlled release of doxorubicin, Nanocarriers for dermal drug delivery: influence of preparation
Carbohydr. Polym. 84 (2011) 1357–1364. method, carrier type and rheological properties, Int. J. Pharm. 437 (2012)
[38] D. Peters, R.H. Muller, C. Keck, Development and physical stability of 83–88.
nanostructured lipid carriers (NLC) in buffer solution, in: Annual Meeting of [62] A.V. Heydenreich, R. Westmeier, N. Pedersen, H.S. Poulsen, H.G. Kristensen,
the American Association of Pharmaceutical Scientists, Washington, DC, Preparation and purification of cationic solid lipid nanospheres – effects on
2011. particle size, physical stability and cell toxicity, Int. J. Pharm. 254 (2003) 83–
[39] M.A. Schubert, C.C. Muller-Goymann, Characterisation of surface-modified 87.
solid lipid nanoparticles (SLN): influence of lecithin and nonionic emulsifier, [63] X. Dong, C.A. Mattingly, M. Tseng, M. Cho, V.R. Adams, R.J. Mumper,
Eur. J. Pharm. Biopharm. 61 (2005) 77–86. Development of new lipid-based paclitaxel nanoparticles using sequential
[40] N. Schöler, C. Olbrich, K. Tabatt, R.H. Müller, H. Hahn, O. Liesenfeld, simplex optimization, Eur. J. Pharm. Biopharm. 72 (2009) 9–17.
Surfactant, but not the size of solid lipid nanoparticles (SLN) influences [64] J.M. Koziara, P.R. Lockman, D.D. Allen, R.J. Mumper, Paclitaxel nanoparticles
viability and cytokine production of macrophages, Int. J. Pharm. 221 (2001) for the potential treatment of brain tumors, J. Control. Release 99 (2004) 259–
57–67. 269.
[41] Z. Magdolenova, D. Bilanicova, G. Pojana, L.M. Fjellsbo, A. Hudecova, K. [65] L. Serpe, M.G. Catalano, R. Cavalli, E. Ugazio, O. Bosco, R. Canaparo, E. Muntoni,
Hasplova, A. Marcomini, M. Dusinska, Impact of agglomeration and different R. Frairia, M.R. Gasco, M. Eandi, G.P. Zara, Cytotoxicity of anticancer drugs
dispersions of titanium dioxide nanoparticles on the human related in vitro incorporated in solid lipid nanoparticles on HT-29 colorectal cancer cell line,
cytotoxicity and genotoxicity, J. Environ. Monit. 14 (2012) 455–464. Eur. J. Pharm. Biopharm. 58 (2004) 673–680.
[42] F. Watari, N. Takashi, A. Yokoyama, M. Uo, T. Akasaka, Y. Sato, S. Abe, Y. [66] X.-G. Zhang, J. Miao, Y.-Q. Dai, Y.-Z. Du, H. Yuan, F.-Q. Hu, Reversal activity of
Totsuka, K. Tohji, Material nanosizing effect on living organisms: non- nanostructured lipid carriers loading cytotoxic drug in multi-drug resistant
specific, biointeractive, physical size effects, J. R. Soc. Interface 6 (2009) S371– cancer cells, Int. J. Pharm. 361 (2008) 239–244.
S388. [67] V.K. Venishetty, R. Komuravelli, M. Kuncha, R. Sistla, P.V. Diwan, Increased
[43] C. Carbone, B. Tomasello, B. Ruozi, M. Renis, G. Puglisi, Preparation and brain uptake of docetaxel and ketoconazole loaded folate grafted solid lipid
optimization of PIT solid lipid nanoparticles via statistical factorial design, nanoparticles, Nanomed. NBM 9 (2013) 111–121.
Eur. J. Med. Chem. 49 (2012) 110–117. [68] L. Wang, M. Li, N. Zhang, Folate-targeted docetaxel-lipid-based-
[44] R. Abbasalipourkabir, A. Salehzadeh, R. Abdullah, Cytotoxicity effect of solid nanosuspensions for active-targeted cancer therapy, Int. J. Nanomed. 7
lipid nanoparticles on human breast cancer cell lines, Biotechnology 10 (2012) 3281–3294.
(2011) 528–533. [69] S.V. Mussi, R.C. Silva, M.C.d. Oliveira, C.M. Lucci, R.B.d. Azevedo, L.A.M.
[45] H. Yang, C. Liu, D. Yang, H. Zhang, Z. Xi, Comparative study of cytotoxicity, Ferreira, New approach to improve encapsulation and antitumor activity of
oxidative stress and genotoxicity induced by four typical nanomaterials: the doxorubicin loaded in solid lipid nanoparticles, Eur. J. Pharm. Sci. 48 (2012)
role of particle size, shape and composition, J. Appl. Toxicol. 29 (2009) 69–78. 282–290.
[46] U. Ruktanonchai, P. Bejrapha, U. Sakulkhu, P. Opanasopit, N. [70] Y.-C. Kuo, C.-T. Liang, Catanionic solid lipid nanoparticles carrying
Bunyapraphatsara, V. Junyaprasert, S. Puttipipatkhachorn, Physicochemical doxorubicin for inhibiting the growth of U87MG cells, Colloids Surf., B:
characteristics, cytotoxicity, and antioxidant activity of three lipid Biointerfaces 85 (2011) 131–137.
nanoparticulate formulations of alpha-lipoic acid, AAPS PharmSciTech 10 [71] P. Zhang, G. Ling, X. Pan, J. Sun, T. Zhang, X. Pu, S. Yin, Z. He, Novel
(2009) 227–234. nanostructured lipid-dextran sulfate hybrid carriers (NLDCs) overcome
[47] U. Ruktanonchai, U. Sakulkhu, P. Bejrapha, P. Opanasopit, N. tumor multidrug resistance of mitoxantrone hydrochloride, Nanomed. NBM
Bunyapraphatsara, V. Junyaprasert, S. Puttipipatkhachorn, Effect of lipid 8 (2012) 185–193.
types on physicochemical characteristics, stability and antioxidant activity of [72] J. Varshosaz, F. Hassanzadeh, H. Sadeghi, M. Shakery, Folate targeted solid
gamma-oryzanol-loaded lipid nanoparticles, J. Microencapsul. 26 (2009) lipid nanoparticles of simvastatin for enhanced cytotoxic effects of
614–626. doxorubicin in chronic myeloid leukemia, Curr. Nanosci. 8 (2012) 249–258.
[48] S. Trombino, R. Cassano, R. Muzzalupo, A. Pingitore, E. Cione, N. Picci, Stearyl [73] J.S. Baek, C.W. Cho, 2-Hydroxypropyl-beta-cyclodextrin-modified SLN of
ferulate-based solid lipid nanoparticles for the encapsulation and paclitaxel for overcoming p-glycoprotein function in multidrug-resistant
stabilization of [beta]-carotene and [alpha]-tocopherol, Colloids Surf., B: breast cancer cells, J. Pharm. Pharmacol. 65 (2012) 72–78.
Biointerfaces 72 (2009) 181–187. [74] X. Lin, R. Gao, Y. Zhang, N. Qi, Y. Zhang, K. Zhang, H. He, X. Tang, Lipid
[49] A. Miglietta, R. Cavalli, C. Bocca, L. Gabriel, M. Rosa Gasco, Cellular uptake and nanoparticles for chemotherapeutic applications: strategies to improve
cytotoxicity of solid lipid nanospheres (SLN) incorporating doxorubicin or anticancer efficacy, Exp. Opin. Drug. Deliv. 9 (2012) 767–781.
paclitaxel, Int. J. Pharm. 210 (2000) 61–67. [75] A.L.R.d. Souza, in: Desenvolvimento de nanopartículas lipídicas sólidas
[50] R.R. Zhu, L.L. Qin, M. Wang, S.M. Wu, S.L. Wang, R. Zhang, Z.X. Liu, X.Y. Sun, contendo praziquantel, Faculdade de Ciências Farmacêuticas, Universidade
S.D. Yao, Preparation, characterization, and anti-tumor property of Estadual Paulista. ‘‘Júlio de Mesquita Filho’’, Araraquara, S. Paulo, Brazil, 2011.
S. Doktorovova et al. / European Journal of Pharmaceutics and Biopharmaceutics 87 (2014) 1–18 17

[76] F. Wan, J. You, Y. Sun, X.-G. Zhang, F.-D. Cui, Y.-Z. Du, H. Yuan, F.-Q. Hu, for Alzheimers disease: preparation, characterization and cytotoxicity
Studies on PEG-modified SLNs loading vinorelbine bitartrate (I): preparation studies, Curr. Nanosci. 5 (2009) 26–32.
and evaluation in vitro, Int. J. Pharm. 359 (2008) 104–110. [102] E.H. Gokce, E. Korkmaz, S. Tuncay-Tanrıverdi, E. Dellera, G. Sandri, M.C.
[77] D.C. Drummond, C.O. Noble, Z. Guo, M.E. Hayes, C. Connolly-Ingram, B.S. Bonferoni, O. Ozer, A comparative evaluation of coenzyme Q10-loaded
Gabriel, B. Hann, B. Liu, J.W. Park, K. Hong, C.C. Benz, J.D. Marks, D.B. Kirpotin, liposomes and solid lipid nanoparticles as dermal antioxidant carriers, Int. J.
Development of a highly stable and targetable nanoliposomal formulation of Nanomed. 2012 (2012) 5109–5117.
topotecan, J Control. Release 141 (2010) 13–21. [103] V. Kakkar, I.P. Kaur, Evaluating potential of curcumin loaded solid lipid
[78] F. Lacoeuille, E. Garcion, J.P. Benoit, A. Lamprecht, Lipid nanocapsules for nanoparticles in aluminium induced behavioral, biochemical and
intracellular drug delivery of anticancer drugs, J. Nanosci. Nanotechnol. 7 histopathological alterations in mice brain, Food Chem. Toxicol. 49 (2011)
(2007) 4612–4617. 2906–2913.
[79] G. Carneiro, E.L. Silva, L.A. Pacheco, E.M. de Souza-Fagundes, N.C. Correa, A.M. [104] S. Doktorovova, A.M. Silva, I. Gaivão, J.P. Teixeira, E.B. Souto, P. Martins-Lopes,
de Goes, M.C. de Oliveira, L.A. Ferreira, Formation of ion pairing as an Comet assay reveals no genotoxicity risk of cationic solid lipid nanoparticles,
alternative to improve encapsulation and anticancer activity of all-trans J. Appl. Toxicol. (2014), http://dx.doi.org/10.1002/jat.2961.
retinoic acid loaded in solid lipid nanoparticles, Int. J. Nanomed. 7 (2012) [105] S. Doktorovová, A.M. Silva, I. Gaivão, C.M. Lopes, E.B. Souto, J.P. Teixeira, P.
6011–6020. Martins-Lopes, Genotoxicity evaluation of cationic solid lipid nanoparticles,
[80] M. Wang, L. Qin, K. Li, R. Zhu, W. Wang, S. Wang, The improvement of the in: International Conference on Environmental and Occupational Health
anticancer effect of a novel compound benzoic acid, 2-hydroxy-, 2-D- 2011, Porto, Portugal, 2011.
ribofuranosylhydrazide (BHR) loaded in solid lipid nanoparticles, AAPS [106] H. Sies, Biochemistry of oxidative stress, Angew. Chem. Int. Ed. 25 (1986)
PharmSciTech 13 (2012) 1348–1354. 1058–1071.
[81] C. Han, C.M. Qi, B.K. Zhao, J. Cao, S.Y. Xie, S.L. Wang, W.Z. Zhou, Hydrogenated [107] Y. Wang, W. Wu, In situ evading of phagocytic uptake of stealth solid lipid
castor oil nanoparticles as carriers for the subcutaneous administration of nanoparticles by mouse peritoneal macrophages, Drug Deliv. 13 (2006) 189–192.
tilmicosin: in vitro and in vivo studies, J. Vet. Pharm. Ther. 32 (2009) 116– [108] J. Koziara, J. Oh, W. Akers, S. Ferraris, R. Mumper, Blood compatibility of cetyl
123. alcohol/polysorbate-based nanoparticles, Pharm. Res. 22 (2005) 1821–1828.
[82] E.H. Gokce, G. Sandri, S. Egrilmez, M.C. Bonferoni, T. Guneri, C. Caramella, [109] A. Jain, A. Agarwal, S. Majumder, N. Lariya, A. Khaya, H. Agrawal, S. Majumdar,
Cyclosporine A-loaded solid lipid nanoparticles: ocular tolerance and in vivo G.P. Agrawal, Mannosylated solid lipid nanoparticles as vectors for site-
drug release in rabbit eyes, Curr. Eye Res. 34 (2009) 996–1003. specific delivery of an anti-cancer drug, J. Control. Release 148 (2010) 359–
[83] A.L.R. Souza, T. Andreani, S. Doktorovova, A.M. Silva, E.B. Souto, M.P.D. 367.
Gremiao, Development of solid lipid nanoparticles for praziquantel delivery: [110] A. Agarwal, S. Majumder, H. Agrawal, S. Majumdar, G.P. Agrawal, Cationized
particle size characterization and cell toxicity assessment, Sci. Pharm. 78 albumin conjugated solid lipid nanoparticles as vectors for brain delivery of
(2010) 576, http://dx.doi.org/10.3797/scipharm.cespt.8.LNT02. an anti-cancer drug, Curr. Nanosci. 7 (2011) 71–80.
[84] Y. Chen, L. Yuan, L. Zhou, Z. Zhang, W. Cao, Q. Wu, Effect of cell-penetrating [111] J.H. Healey, Editorial: the imperative to authenticate cell lines, Clin. Orthop.
peptide-coated nanostructured lipid carriers on the oral absorption of Relat. Res. 468 (2010) 3413–3414.
tripterine, Int. J. Nanomed. 7 (2012) 4581–4591. [112] J. Fangueiro, E. Gonzalez-Mira, P. Martins-Lopes, M. Egea, M. Garcia, S. Souto,
[85] K. Teskac, J. Kristl, The evidence for solid lipid nanoparticles mediated cell E. Souto, A novel lipid nanocarrier for insulin delivery: production,
uptake of resveratrol, Int. J. Pharm. 390 (2010) 61–69. characterization and toxicity testing, Pharm. Dev. Technol. 18 (2013) 545–
[86] F. Li, Y. Weng, L. Wang, H. He, J. Yang, X. Tang, The efficacy and safety of 549.
bufadienolides-loaded nanostructured lipid carriers, Int. J. Pharm. 393 (2010) [113] C. Olbrich, R.H. Müller, K. Tabatt, O. Kayser, C. Schulze, R. Schade, Stable
204–212. biocompatible adjuvants – a new type of adjuvant based on solid lipid
[87] E.H. Gokce, E. Korkmaz, E. Dellera, G. Sandri, M.C. Bonferoni, O. Ozer, nanoparticles: a study on cytotoxicity, compatibility and efficacy in chicken,
Resveratrol-loaded solid lipid nanoparticles versus nanostructured lipid ATLA 30 (2002) 443–458.
carriers: evaluation of antioxidant potential for dermal applications, Int. J. [114] Z.H. Xu, L.L. Chen, W.W. Gu, Y. Gao, L.P. Lin, Z.W. Zhang, Y. Xi, Y.P. Li, The
Nanomed. 7 (2012) 1841–1850. performance of docetaxel-loaded solid lipid nanoparticles targeted to
[88] S. Wang, T. Chen, R. Chen, Y. Hu, M. Chen, Y. Wang, Emodin loaded solid lipid hepatocellular carcinoma, Biomaterials 30 (2009) 226–232.
nanoparticles: preparation, characterization and antitumor activity studies, [115] S. Martins, I. Tho, I. Reimold, G. Fricker, E. Souto, D. Ferreira, M. Brandl, Brain
Int. J. Pharm. 430 (2012) 238–246. delivery of camptothecin by means of solid lipid nanoparticles: formulation
[89] J. Shen, Y. Deng, X. Jin, Q. Ping, Z. Su, L. Li, Thiolated nanostructured lipid design, in vitro and in vivo studies, Int. J. Pharm. 439 (2012) 49–62.
carriers as a potential ocular drug delivery system for cyclosporine A: [116] M.L. Bondi, E.F. Craparo, G. Giammona, M. Cervello, A. Azzolina, P. Diana, A.
improving in vivo ocular distribution, Int. J. Pharm. 402 (2010) 248–253. Martorana, G. Cirrincione, Nanostructured lipid carriers-containing
[90] R.R. Patlolla, M. Chougule, A.R. Patel, T. Jackson, P.N.V. Tata, M. Singh, anticancer compounds: preparation, characterization, and cytotoxicity
Formulation, characterization and pulmonary deposition of nebulized studies, Drug Deliv. 14 (2007) 61–67.
celecoxib encapsulated nanostructured lipid carriers, J. Control. Release 144 [117] N. Pedersen, S. Hansen, A.V. Heydenreich, H.G. Kristensen, H.S. Poulsen, Solid
(2010) 233–241. lipid nanoparticles can effectively bind DNA, streptavidin and biotinylated
[91] R. Lopes, C.V. Eleuterio, L.M.D. Gonçalves, M.E.M. Cruz, A.J. Almeida, Lipid ligands, Eur. J. Pharm. Biopharm. 62 (2006) 155–162.
nanoparticles containing oryzalin for the treatment of leishmaniasis, Eur. J. [118] L. Hu, Y. Jia, WenDing, Preparation and characterization of solid lipid
Pharm. Sci. 45 (2012) 442–450. nanoparticles loaded with epirubicin for pulmonary delivery, Pharmazie 65
[92] N.B. Wolf, S. Küchler, M.R. Radowski, T. Blaschke, K.D. Kramer, G. Weindl, B. (2010) 585–587.
Kleuser, R. Haag, M. Schäfer-Korting, Influences of opioids and nanoparticles [119] E.H. Gokce, G. Sandri, M.C. Bonferoni, S. Rossi, F. Ferrari, T. Güneri, C.
on in vitro wound healing models, Eur. J. Pharm. Biopharm. 73 (2009) 34–42. Caramella, Cyclosporine A loaded SLNs: evaluation of cellular uptake and
[93] R.H. Müller, S. Gohla, C.M. Keck, State of the Artof nanocrystals-special corneal cytotoxicity, Int. J. Pharm. 364 (2008) 76–86.
features, production, nanotoxicology aspects & intracellular delivery, Eur. J. [120] L. Zhou, Y. Chen, Z. Zhang, J. He, M. Du, Q. Wu, Preparation of tripterine
Pharm. Biopharm. 78 (2011) 1–9. nanostructured lipid carriers and their absorption in rat intestine, Pharmazie
[94] S.E.A. Gratton, P.A. Ropp, P.D. Pohlhaus, J.C. Luft, V.J. Madden, M.E. Napier, J.M. 67 (2012) 304–310.
DeSimone, The effect of particle design on cellular internalization pathways, [121] T. Ramasamy, U.S. Khandasami, H. Ruttala, S. Shanmugam, Development of
Proc. Natl. Acad. Sci. U. S. A. 105 (2008) 11613–11618. solid lipid nanoparticles enriched hydrogels for topical delivery of anti-fungal
[95] C. Olbrich, U. Bakowsky, C.-M. Lehr, R.H. Müller, C. Kneuer, Cationic solid– agent, Macromol. Res. 20 (2012) 682–692.
lipid nanoparticles can efficiently bind and transfect plasmid DNA, J. Control. [122] E. Vighi, M. Montanari, M. Hanuskova, V. Iannuccelli, G. Coppi, E. Leo, Design
Release 77 (2001) 345–355. flexibility influencing the in vitro behavior of cationic SLN as a nonviral gene
[96] H.R. Kim, I.K. Kim, K.H. Bae, S.H. Lee, Y. Lee, T.G. Park, Cationic solid lipid vector, Int. J. Pharm. 440 (2013) 161–169.
nanoparticles reconstituted from low density lipoprotein components for [123] D. Ling, B.-c. Bae, W. Park, K. Na, Photodynamic efficacy of photosensitizers
delivery of siRNA, Mol. Pharm. 5 (2008) 622–631. under an attenuated light dose via lipid nano-carrier-mediated nuclear
[97] T.C. Long, N. Saleh, R.D. Tilton, G.V. Lowry, B. Veronesi, Titanium dioxide (P25) targeting, Biomaterials 33 (2012) 5478–5486.
produces reactive oxygen species in immortalized brain microglia (BV2): [124] M. Wang, L. Qin, K. Li, R. Zhu, W. Wang, S. Wang, The improvement of the
implications for nanoparticle neurotoxicity, Environ. Sci. Technol. 40 (2006) anticancer effect of a novel compound benzoic acid, 2-hydroxy-, 2-D-
4346–4352. ribofuranosylhydrazide (BHR) loaded in solid lipid nanoparticles, AAPS
[98] X. Chen, Z. Zhong, Z. Xu, L. Chen, Y. Wang, 20 ,70 -Dichlorodihydrofluorescein as PharmSciTech 13 (2012) 1348–1354.
a fluorescent probe for reactive oxygen species measurement: forty years of [125] S. Taveira, F.A. de Campos, L.M.P. jo, D.C.A.S. de Santana, A. Nomizo, L.A.P. de
application and controversy, Free Radical Res. 44 (2011) 587–604. Freitas, R.F.V. Lopez, Development of cationic solid lipid nanoparticles with
[99] M.M. Tarpey, D.A. Wink, M.B. Grisham, Methods for detection of reactive factorial design-based studies for topical administration of doxorubicin, J.
metabolites of oxygen and nitrogen: in vitro and in vivo considerations, Am. J. Biomed. Nanotechnol. 8 (2012) 219–228.
Physiol. Regul. Integr. Comp. Physiol. 286 (2004) R431–R444. [126] R. Asasutjarit, S.-I. Lorenzen, S. Sirivichayakul, K. Ruxrungtham, U.
[100] P. Picone, M.L. Bondi, G. Montana, A. Bruno, G. Pitarresi, G. Giammona, M. Di Ruktanonchai, G. Ritthidej, Effect of solid lipid nanoparticles formulation
Carlo, Ferulic acid inhibits oxidative stress and cell death induced by Ab compositions on their size, zeta potential and potential for in vitro pHIS-HIV-
oligomers: improved delivery by solid lipid nanoparticles, Free Radical Res. hugag transfection, Pharm. Res. 24 (2007) 1098–1107.
43 (2009) 1133–1145. [127] W.H. Kong, K. Park, M.-Y. Lee, H. Lee, D.K. Sung, S.K. Hahn, Cationic solid lipid
[101] M.L. Bondi, G. Montana, E.F. Craparo, P. Picone, G. Capuano, M.D. Carlo, G. nanoparticles derived from apolipoprotein-free LDLs for target specific
Giammona, Ferulic acid-loaded lipid nanostructures as drug delivery systems systemic treatment of liver fibrosis, Biomaterials 34 (2013) 542–551.
18 S. Doktorovova et al. / European Journal of Pharmaceutics and Biopharmaceutics 87 (2014) 1–18

[128] J. Miao, Y.-Z. Du, H. Yuan, X.-G. Zhang, F.-Q. Hu, Drug resistance reversal [134] A. Chuan, Y. Li, L. Yang, X. Sun, Q. Zhang, T. Gong, Z. Zhang, Enhanced
activity of anticancer drug loaded solid lipid nanoparticles in multi-drug rifampicin delivery to alveolar macrophages by solid lipid nanoparticles, J.
resistant cancer cells, Colloid Surf., B 110 (2013) 74–80. Nanopart. Res. 15 (2013) 1634, http://dx.doi.org/10.1007/s11051-013-1634-
[129] S.M. Martins, B. Sarmento, C. Nunes, M. Lucio, S. Reis, D.C. Ferreira, 1.
Brain targeting effect of camptothecin-loaded solid lipid nanoparticles [135] A. Khajavinia, J. Varshosaz, A. Dehkordi, Targeting etoposide to acute
in rat after intravenous administration, Eur. J. Pharm. Biopharm. 85 (2013) myelogenous leukaemia cells using nanostructured lipid carriers coated
488–502. with transferrin, Nanotechnology 23 (2012) 405101.
[130] M. Videira, A.J. Almeida, A. Fabra, Preclinical evaluation of a pulmonary [136] C.W. How, R. Abdullah, R. Abbasalipourkabir, Characterization and
delivered paclitaxel-loaded lipid nanocarrier antitumor effect, Nanomedicine cytotoxicity of nanostructured lipid carriers formulated with olive oil,
8 (2012) 1208–1215. hydrogenated palm oil and Polysorbate 80, IEEE Trans. Nanobiosci. 12
[131] E. Vighi, M. Montanari, M. Hanuskova, V. Iannuccelli, G. Coppi, E. Leo, Design (2012) 72–78.
flexibility influencing the in vitro behavior of cationic SLN as a nonviral gene [137] G. Carneiro, E.L. Silva, L.A. Pacheco, E.M. de Souza-Fagundes, N.C. Correa, A.M.
vector, Int. J. Pharm. 440 (2013) 161–169. de Goes, M.C. de Oliveira, L.A. Ferreira, Formation of ion pairing as an
[132] M.Á. Aznar, B. Lasa-Saracíbar, A. Estella-Hermoso de Mendoza, M.J. Blanco- alternative to improve encapsulation and anticancer activity of all-trans
Prieto, Efficacy of edelfosine lipid nanoparticles in breast cancer cells, Int. J. retinoic acid loaded in solid lipid nanoparticles, Int. J. Nanomed. 7 (2012)
Pharm. 454 (2013) 720–726. 6011–6020.
[133] S.V. Mussi, R.C. Silva, M.C.d. Oliveira, C.M. Lucci, R.B.d. Azevedo, L.A.M. [138] C. Vitorino, J. Almeida, L.M. Gonçalves, A.J. Almeida, J.J. Sousa, A.A.C.C. Pais,
Ferreira, New approach to improve encapsulation and antitumor activity of Co-encapsulating nanostructured lipid carriers for transdermal application:
doxorubicin loaded in solid lipid nanoparticles, Eur. J. Pharm. Sci. 48 (2012) from experimental design to the molecular detail, J. Control. Release 167
282–290. (2013) 301–314.

S-ar putea să vă placă și