Sie sind auf Seite 1von 20

Advances in Colloid and Interface Science 205 (2014) 187–206

Contents lists available at ScienceDirect

Advances in Colloid and Interface Science


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

Niosomes from 80s to present: The state of the art


Carlotta Marianecci a,1, Luisa Di Marzio b,1, Federica Rinaldi a, Christian Celia b,c, Donatella Paolino d,
Franco Alhaique a, Sara Esposito b, Maria Carafa a,⁎
a
Department of Drug Chemistry and Technologies, University “Sapienza” of Rome, Rome, Italy
b
Department of Pharmacy, University “G. d'Annunzio”, Chieti, Italy
c
Department of Nanomedicine, The Methodist Hospital Research Institute, Houston, USA
d
Department of Health Sciences, University “Magna Graecia” of Catanzaro, University Campus “S. Venuta”, Catanzaro, Italy

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

Available online 11 December 2013 Efficient and safe drug delivery has always been a challenge in medicine. The use of nanotechnology, such as the
development of nanocarriers for drug delivery, has received great attention owing to the potential that
Keywords: nanocarriers can theoretically act as “magic bullets” and selectively target affected organs and cells while sparing
Surfactants normal tissues. During the last decades the formulation of surfactant vesicles, as a tool to improve drug delivery,
Vesicular carriers brought an ever increasing interest among the scientists working in the area of drug delivery systems. Niosomes
Niosomes
are self assembled vesicular nanocarriers obtained by hydration of synthetic surfactants and appropriate
Drug delivery
Drug targeting
amounts of cholesterol or other amphiphilic molecules. Just like liposomes, niosomes can be unilamellar or
multilamellar, are suitable as carriers of both hydrophilic and lipophilic drugs and are able to deliver drugs to
the target site. Furthermore, niosomal vesicles, that are usually non-toxic, require less production costs and are
stable over a longer period of time in different conditions, so overcoming some drawbacks of liposomes.
The niosome properties are specifically dictated by size, shape, and surface chemistry which are able to modify
the drug's intrinsic pharmacokinetics and eventual drug targeting to the areas of pathology.
This up-to-date review deals with composition, preparation, characterization/evaluation, advantages, disadvan-
tages and application of niosomes.
© 2013 Elsevier B.V. All rights reserved.

Contents

1. Introduction . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 188
2. Factors leading to niosome formation . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 189
3. Niosomal formulation . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 190
3.1. Non-ionic surfactants . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 190
3.2. Cholesterol . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 190
3.3. Charged molecules . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 191
4. Issues related to niosomal formulation . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 191
4.1. Vesicle preparation . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 191
4.1.1. Proniosome . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 192
4.1.2. Heating method (HM) . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 192
4.1.3. Supercritical carbon dioxide fluid (scCO2) . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 192
4.1.4. Membrane contactor . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 192
4.1.5. Microfluidic hydrodynamic focusing . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 192
4.2. Vesicle purification . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 193
4.2.1. Dialysis . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 193
4.2.2. Gel filtration . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 193
4.2.3. Centrifugation/ultracentrifugation . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 193

⁎ Corresponding author at: Department of Chemistry and Technologies, University of Rome “Sapienza”, P.le A. Moro 5, 00185 Roma, Italy. Tel.: +39 0649913603; fax: +39 0649913133.
E-mail address: maria.carafa@uniroma1.it (M. Carafa).
1
These authors equally contributed to the work.

0001-8686/$ – see front matter © 2013 Elsevier B.V. All rights reserved.
http://dx.doi.org/10.1016/j.cis.2013.11.018
188 C. Marianecci et al. / Advances in Colloid and Interface Science 205 (2014) 187–206

4.3.Vesicle characterization . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 194


4.3.1. Vesicle size . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 194
4.3.2. ζ-potential and surface properties . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 194
4.3.3. Bilayer characterization . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 194
4.3.4. Vesicle stability . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 194
4.3.5. Entrapment efficiency . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 194
4.3.6. pH-sensitivity assessment . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 195
5. Niosomes in drug delivery and targeting . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 195
5.1. Dermal and transdermal delivery . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 195
5.2. Ocular delivery . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 196
5.3. Oral delivery . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 197
5.4. Pulmonary delivery . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 198
5.5. Parenteral delivery . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 199
5.6. Gene delivery . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 201
5.7. Diagnostics/theranostics . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 201
6. Natural products delivery . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 201
7. Niosomes vs liposomes . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 202
8. Niosome-based patents and pre-clinical and clinical trials . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 203
9. Concluding remarks and perspectives . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 203
References . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 204

1. Introduction other therapeutic agents specifically into target sites. Many of the
conventional nano DDS (e.g. liposomes, micelles, and polymer-based
The use of both conventional and new drug delivery systems (DDS) nanodevices) have reached the late stages of development, and some
allows to face the major issues in drug release: (1) unfavorable pharma- of them were approved. Over the last two decades, the development
cokinetics and biodistribution which lead to unwanted side effects of synthesis and characterization techniques including the ability to
(e.g. chemotherapy), (2) early drug degradation in the bloodstream by manipulate molecules and supramolecular structures for beneficial
reticulo-endothelial system (RES), and (3) inefficient uptake at target functions blossomed into innovative-engineered nanomaterials suit-
sites that leads to low drug efficacy. In this sense, nanocarriers offer an able for biopharmaceutical/therapeutic applications. Another important
innovative approach to drug delivery, providing a range of features development is that nanocarriers are becoming highly integrated and
including cargo protection and increased dose delivery to target sites. multifunctional so as to include a range of applications such as on-
Actually, the complexity of human diseases may lead to ineffective demand release, specific tissue/cell type targeting, in vivo imaging and
in vivo drug treatments, consequently the development of biologically diagnosis, and photothermal treatment [1–4].
functional nanocarriers that incorporate drugs to selectively bind and The ultimate goal of nano DDS is to generate therapeutically and
target specific cells becomes crucial. There have been many attempts clinically useful formulations for disease treatments in patients. While
to functionalize and design nanocarriers so as to increase their efficacy; actively studied in academia and industry, the pursuit of optimized
nevertheless, although the criteria for a successful nanocarrier are quite nanocarriers is in continuous evolution and the topic requires innova-
numerous, only few nanocarriers have actually shown significant clinical tion and skill in order to translate them into human trials. A successful
potential. Inspired by the unique properties of non-ionic surfactant ves- nanocarrier should ideally meet the following requirements:
icles (NSVs), our review expands on the versatility and flexibility of NSVs (1) formulation with biocompatible/biodegradable/bioexcretable
and how such nanostructures can be used for therapeutic purposes. materials, (2) high drug and cargo loading capacity, (3) site-specific
Due to the rapid growth in nanotechnology, it can be asserted that delivery mechanism to avoid normal cells and tissues, (4) zero or negli-
the study of drug delivery and disease treatment has been revolution- gible premature drug release, and (5) controlled release mechanism
ized. By molding nanomaterials into vesicles, numerous nanocarriers to provide an effective dose to the target site. To date, there is a very
(Fig. 1) have been developed to securely deliver drugs and various limited number of nanocarriers which can achieve such prerequisites.

Liposome Micelle Dendrimer Polymeric Nanoparticle

Fe3O4

Carbon Nanotube Quantum Dots Iron Oxide Gold Nanoparticle Mesoporous Silica

Fig. 1. The leading nanocarriers for drug delivery: the top row shows the representative conventional nanocarriers while the bottom row shows novel inorganic nanocarriers.
C. Marianecci et al. / Advances in Colloid and Interface Science 205 (2014) 187–206 189

At the present state of the art, numerous scientists throughout the The main factors that play a role in amphiphile self-assembling
world are involved in the study of vesicles and other colloidal structures are thermodynamic and physicochemical parameters such as the
as carriers for drug delivery. hydrophilic-lipophilic balance (HLB) and the geometric feature of the
Liposomes, described by Bangham in 1965, have been prepared with amphiphilic molecules; but several other factors are relevant for vesicle
a variety of phospholipids and have been extensively studied as drug formation and properties and must be taken into consideration: in
carriers [5–7]. particular aqueous interlayer, lipid chain-length, chain-packing and
Although phospholipids are biodegradable and non-toxic amphi- membrane asymmetry. The energy required to form vesicles with am-
philes, problems arise in practical applications of liposomes, because phiphilic molecules has three contributors related to the surface energy,
of the low physical and chemical stability of aqueous suspensions of the mechanical energy due to overpressure and the chemical potential
this type of vesicles. For these reasons, other amphiphilic molecules, excess. The association of non-ionic surfactant monomers into vesicles
alternative to phospholipids, have been studied to obtain vesicular upon hydration is a result of the high interfacial tension between
carriers: non-ionic surfactants [8,9]. water and hydrocarbon portion (or any other hydrophobic group) of
Vesicles prepared by surfactants are known as niosomes or NSVs. the amphiphile which causes the association of these groups. At the
Niosomes are similar, in terms of structure and physical properties, to same time the steric, hydrophilic, and/or ionic repulsion among the
liposomes [8]. They are also prepared, as unilamellar or multilamellar head groups ensures that these groups are in contact with water. Of
vesicles, following the same procedures and under the same variety of course also monomer concentration and temperature play an important
conditions. role in vesicle formation.
The research interest in niosomal formulations is recently widening Thermodynamically this self-assembly must contend against a
because niosomes are able to overcome some disadvantages associated negative entropy component (ΔS) and the reduction in free energy
with liposomes, surfactants are easily derivatized and give a higher (ΔG) and is only achieved by the favorable enthalpy (ΔH) contribu-
versatility to the vesicular structure and moreover they have lower tion arising from the van der Waals attractions, the hydrophobic
costs than phospholipids. forces, the hydrogen bond formation and the screened electrostatic
The self-assembly of non-ionic surfactant into vesicles was first interactions.
reported in the seventies by researchers in the cosmetic industry [10]. As above pointed out, vesicle formation may depend on the HLB
Since then an ever increasing interest has been exhibited on the applica- value; thus the guidance offered by the HLB number is useful in the
tion of niosomes (Fig. 2) in the field of pharmaceutics, cosmetics and evaluation of new classes of compounds for their vesicles forming ability.
food industry, leading to the publication of more than 1200 research ar- With the sorbitan monostearate surfactants (Sp), an HLB number
ticles, about 200 patents and 6 clinical trials from 1980 (from SciFinder). between 4 and 8 was found to be compatible with vesicle formation
The aim of this paper is to present an overview on niosome prepara- [8]. On the other hand, it was actually shown that, owing to their high
tion and characterization as well as a description of their applications, aqueous solubility, hydrophilic surfactants cannot form free hydrated
with particular attention to more recent pharmaceutical formulations. units (vesicles) but these free units rather aggregate and coalesce to
Considering that this is a fast-moving research field, all aspects of form lamellar structures [15,16]. Consequently, niosomes are not gener-
niosomal applications cannot be included or discussed. This review ally formed with surfactants with a HLB value between 14 and 17 [17]
will provide an overview on the increasing interest on niosomes in the and the addition of cholesterol plays a fundamental role in vesicle
field of disease diagnosis and therapy as well as in the cosmetic field. formation with surfactants with a HLB value around 10 [18].
The geometric feature of the single vesicle-forming unit may be
another important factor in determining the type of aggregate formed
2. Factors leading to niosome formation in aqueous environments [19]. To explain the behavior of non-ionic
surfactants in water, a hydrophobic effect was proposed several years
The self-assembly of amphiphilic molecules into closed bilayers, ago, as the essential strength leading to an enhancement of global
both in the case of liposomes and niosomes, is not spontaneous but it system-free energy. Shapes of the spontaneously formed association
involves some input of energy, for instance by means of physical colloids can be predicted with considerable certainty using nominal
shaking (hand-shaking, ultrasound, heat, etc.) [11,12]. Moreover, ther- geometric parameters of the surfactant molecule.
modynamically stable vesicles are formed only in presence of appropri- The critical packing parameter (CPP also named as Ps) was discussed
ate mixtures of surfactants and charge inducing agents. Although by Israelachvili et al. in 1976 [20] and defined by relation:
previous studies showed that niosomes have a higher resistance to
micellar solubilization than liposomes [13,14], similar factors lead to
V
the formation of both types of vesicles. CPP ¼ ð1Þ
a0 lc

where:
– V is the tail volume of the molecule,
– a0 is the surface area per molecule at the hydrocarbon–water inter-
face,
– lc is the critical, i.e. the greatest, span of a fluid molecular chain in an
aggregate.
According to the CPP value, the shape and size of the equilibrium
aggregate should evolve from spherical micelles (CPP ≤ 1/3) to cylin-
drical micelles (1/3 ≤ CPP ≤ 1/2), bilayers (1/2 ≤ CPP ≤ 1) or inverse
micelles (CPP N 1) (Fig. 3).
The influence of polar head surface, a0, on the ability to form vesicles
was confirmed by a comparison between Polysorbates (Tw) 21 and 20
bearing the same alkyl chain (dodecanoic acid) but different hydrophilic
Fig. 2. General stage of the development of niosomal carriers in pharmaceutical applications head groups (Tw21:4 PEG units; Tw20:20 PEG units) [21], thus
from 1980 (from SciFinder). influencing also the HLB value (Tw20 HLB: 16.7; Tw21 HLB: 13.3).
190 C. Marianecci et al. / Advances in Colloid and Interface Science 205 (2014) 187–206

Fig. 3. The relationship between the CPP and the morphology of self-assembled amphiphilic molecules.

Recently it was reported by Cevc [22] that scrutinizing amphiphile Alkyl ether surfactants can be broadly divided into two classes based
aggregation in terms of the popular molecular descriptors (esp. the on the nature of the hydrophilic head group: alkyl ethers in which the
CPP or HLB number) could be “too static” to foretell reliably and quanti- hydrophilic head group consists of repeating glycerol subunits, related
tatively bilayer vesicle formation. Consequently, a different predictor isomers or larger sugar molecules and those in which the hydrophilic
was proposed: the effective area per lipid chain (cross-section of a head group consists of repeating ethylene oxide subunits.
“tail”, Ac), which seems to correlate, quasi-exponentially, with the The first niosomes were formulated using cholesterol and single-
ease of bilayer vesicle formation and bilayer deformability. chain surfactants such as alkyl (usually from C12 to C18) oxyethylenes
According to this parameter, an Ac value above 0.43 nm2 will identify [8]. Polyglycerol monoalkyl ethers and polyoxylate analogs are the
a micellar structure; a value slightly below this limiting area should most widely used single-chain surfactants. Ether surfactants, suitable
imply bilayer vesicle formation. On the other side, amphiphiles with for noisome formulations are composed, in their lipophilic moiety, of
an appreciably smaller area per chain, Ac ≪ 0.43 nm2, prefer to form monoalkyl or dialkyl chains. The latest ones, being similar to phospho-
multilamellar structures. lipids, and possess higher encapsulation efficiency. Esther type amphi-
It must be underlined that the assigning of an absolute value to philic surfactants are also used for niosome formulation. They are
geometric or molecular parameters is at least misleading, if not wrong. degraded by estherases to triglycerides and fatty acids. Although these
This is due to such molecular and geometric descriptors sensitivity to types of surfactants are less stable than ether type ones, they are usually
the boundary conditions and to the different possibilities to combine less toxic. Finally, also glucosides of myristil, cethyl and stearyl alcohols
components and/or experimental conditions in surfactant vesicle can be used to form niosomes.
preparation. While the number of hydrophobic permutations is at present quite
It should be more correct to consider all reported parameters as limited, there has been a wide variety of hydrophilic head groups in
adjustable and it can consequently be useful to combine them in vesicle forming surfactants, and the design of new molecules is in con-
predicting amphiphilic molecules capability to aggregate into bilayer tinuous evolution, according to the specific vesicle structure that are
vesicles. needed. Some recent studies report the synthesis and the characteriza-
tion of new surfactants with peculiar physical–chemical properties,
3. Niosomal formulation showing promising applications in the field pharmaceutical colloid
science. As an example, a new non-ionic surfactant molecule, in which
The efficacy of a drug delivery system is strictly dependent on the hydrophilic region consists of azacrown ether units, was synthesized
its components, that should be characterized in terms of physical– by Muzzalupo and co-workers. This compound, known as bola-
chemical and clinical properties needed for the formation of niosomal surfactant, is composed of two identical azacrown ether units, as polar
systems with specific characteristics, together with biocompatibility heads, linked to a long alkyl chain [29–31]. The authors demonstrated
and relevance in clinics. The basic components of niosomes include that the bola-surfactant is able to assemble in colloidal structures if
non-ionic surfactants, hydration medium and lipids such as cholesterol. associated with cholesterol or other amphiphilic molecules [30].

3.1. Non-ionic surfactants


3.2. Cholesterol
Just as ionic surfactants, non-ionic surfactants are amphiphilic (or
amphipathic) molecules that have two distinct regions in their chemical It is well known that cholesterol (CHOL) influences the physical
structure, one of which is water-liking or hydrophilic and the other is properties and structure of niosomes because of its interaction with
water-hating or hydrophobic. The two portions of such molecules may non-ionic surfactants. Several surfactants form vesicles only after
be linked by ether, amide or ester bonds. CHOL addition (up to 30–50 mol%). The amount of CHOL to be added
Non-ionic surfactant vesicles can be prepared from different types of depends on the HLB value of the surfactants. As the HLB value increases
molecules, such as: amino acids, fatty acids, amides, alkyl esters and above 10, it is necessary to increase the CHOL concentration in order to
alkyl ether surfactants, the last one being mostly employed for such compensate the effect of the larger head groups on the critical packing
purposes [23–28]. parameter (CPP), previously discussed [32,33].
C. Marianecci et al. / Advances in Colloid and Interface Science 205 (2014) 187–206 191

hydrophilic within the obtained niosomes correspondingly increased,


regardless of the type of stabilizer (Dicetyl phosphate or Solutol HS
15) [44]. Furthermore, CHOL stabilizes niosomes towards destabilizing
effects of plasma and serum components and decreases the permeabil-
ity of vesicles to entrapped solute thus inhibiting leakage [11,45].
According to the above reported results, the still controversial role of
CHOL is evident. The opportunity to add CHOL, and its amounts, needs
to be evaluated case by case, depending on the physical–chemical char-
Tween® 20 acteristic of surfactants and loaded drugs.
x+y+z+w = 20
3.3. Charged molecules
Fig. 4. Possible hydrogen bonding interaction between the 3-OH group of the cholesterol
and the carbonyl oxygen and also weaker interaction at oxygen of the ester bond. One of the methods used to stabilize niosomes is based on the addi-
tion of a charged molecule to the bilayer. Dicetyl phosphate (DCP) and
phosphatidic acid are known as negatively charged molecules, while
The water soluble detergent Tw20 (HLB value = 16.7) forms stable stearylamine (SA) and cetylpyridinium chloride are positively charged
non-ionic surfactant vesicles in the presence of equimolar CHOL con- molecules which are both commonly used for preventing aggregation
centration because in absence of CHOL it would be well hydrated and of niosomes [9]. Normally, the charged molecule is added in niosomal
can be found as free monomers in solution (CMC = 60 mg/l, in water formulation in an amount of 2.5–5 mol% because a higher concentration
at 20 °C) [34–37]. It was suggested that an interaction occurs between of charged molecules could inhibit the formation of niosomes [46,47]. It
the hydrophobic portion of the amphiphile (phospholipid/surfactant) is widely reported that zeta (ζ)-potentials over |30| mV are necessary
next to head group and the 3-OH group of CHOL in vesicular systems for full electrostatic stabilization; potentials between |5| and |15| mV
at an equimolar ratio (CHOL:amphiphile) (Fig. 4) [38] and this interac- represent the region of limited flocculation; while between |5| and |3|
tion could explain the effect of CHOL on the formation and hydration mV correspond to the maximum flocculation. Thus, particle aggregation
behavior of Tw20 niosomal membranes that were found to coexist is less likely to occur for charged particles (high ζ-potential) due to their
with unsolubilized CHOL crystals [39]. electric repulsion. However, this rule cannot strictly be applied for
The role of CHOL in the surfactant vesicle formation is also supported systems containing steric stabilizers, because the adsorption of such
by the work of Manconi et al. [40]. In this study it was reported that a steric stabilizers decreases the ζ-potential because of the shift in the
hydrophilic non-ionic surfactant, decylpolyglucoside, is able to form shear plane of the particle. Moreover, the strong dependence of the
stable and spherical vesicles only in the presence of high amounts of ζ-potential on the ions present in the medium should be taken into
CHOL. These results also suggest that CHOL increases the cohesion account [48]. Charged molecules can be also added to prepare charged
among the non-polar portions of bilayer, thus improving the cohesion niosomes useful for an increase of drug encapsulation efficiency [49],
of niosomal membrane [40]. for skin permeation enhancement [50] and for hybrid niosomal com-
The effect of CHOL in amphiphile bilayers is mostly the modulation plex formation [51], (Fig. 5).
of their mechanical strength and their permeability to water [41,42],
properties that become particularly important under severe stress 4. Issues related to niosomal formulation
conditions (e.g. in vivo) [42], nevertheless it should be pointed that pub-
lished results are not always in agreement. The inclusion of cholesterol 4.1. Vesicle preparation
was successfully used by Manosroi et al. (2003) to increases the hydro-
dynamic diameter and the entrapment efficiency of niosomes prepared Numerous methods for non-ionic surfactant vesicle preparation are
by various non-ionic surfactants [43]. reported, however only some of these are actually used to manufacture
On the other side, it was recently observed that by decreasing liposomes and have been adapted/used as such. Unlike liposomes, the
the molar ratio of cholesterol to Sp 60, the entrapment of a model niosome preparation is usually easier given to the surfactant resistance

A B

Fig. 5. The average hydrodynamic diameter 2R and electrophoretic mobility course of polylysine/niosome structure as a function of the positive/negative charge ratio (A). Schematic
representation of niosomal/polyelectrolite «patchwork like» interaction (B).
192 C. Marianecci et al. / Advances in Colloid and Interface Science 205 (2014) 187–206

against air oxidation. The hydrating temperatures used to make 4.1.2. Heating method (HM)
niosomes should usually be above the gel to liquid phase transition tem- The heating method developed by Mozafari to produce vesicle
perature of the system. The formation of vesicular assemblies requires involves the hydration of an amphipathic molecule in an aqueous solu-
the input of some form of energy [52] and all the experimental methods tion containing 3 vol.% polyol at high temperature. This is a non-toxic,
surveyed consist of the hydration of a mixture of the surfactant: lipid at scalable and one-step method [57].
elevated temperature followed by optional size reduction to obtain a
colloidal dispersion. Moreover, the elevated temperature used does 4.1.3. Supercritical carbon dioxide fluid (scCO2)
not represent an issue, because non-ionic surfactants are more stable In the last years a new method, the supercritical reverse phase
than phospholipids. Furthermore the hydrating temperature must be evaporation (scRPE) method, for the preparation of niosomes using
chosen evaluating drug melting point. Many techniques such as the supercritical fluids has been described by Manosroi [58]. The main
thin film hydration, the organic solvent injection method, the reverse advantages of this method are: the use of no inflammable, volatile,
phase evaporation method and the bubble method have been previous- and non-toxic organic solvent, one step and easy scale-up production
ly described for the preparation of niosomes [9,53] (Table 1). of surfactant vesicles. However, the niosomes prepared with the scCO2
Below the more recent and innovative methods for niosome prepa- method have large unilamellar structure with size between 100 and
ration are reported: 440 nm. A smaller size distribution of the vesicles could be obtained
by the combination of the scCO2 technique with other methods such
as sonication or polycarbonate extrusion.
4.1.1. Proniosome
Proniosome-produced niosomes involve coating a water-soluble 4.1.4. Membrane contactor
carrier (e.g. sucrose stearate, maltodextrin or mannitol) with non- The scaling-up ability of niosome production, from laboratory scale
ionic surfactants. The result of the coating process is a dry formulation using a syringe-pump device to a pilot scale using the membrane
in which each water-soluble particle is covered with a thin film of dry contactor module (Fig. 6), was reported by Pham [59].
surfactant. This preparation is termed “Proniosomes” [54,55].
Important advantages of proniosome-derived niosome are the 4.1.5. Microfluidic hydrodynamic focusing
reduction of niosome physical instability such as aggregation, fusion Conventional bulk method of niosome preparation requires bulk
and leaking and the increase of drug entrapment efficiency [56]. mixing of two liquid phases, which is time-consuming and not well-
In addition, proniosomes are obtained as a dry power, so further controlled. Such mixing conditions often lead to large niosomes with
processing is possible. To provide convenient unit dosing, the high size polydispersity, thus affecting the consistency of niosome
proniosome powder may be processed to make beads, tablets, or cap- dosage or imaging quality. The new method of niosome by microfluidic
sules. The proniosome powder can be implemented in a ‘point-of-use’ hydrodynamic (Fig. 7) leads to improve the size and size distributions of
application. niosomes, by taking advantage of the rapid and controlled mixing of

Table 1
Niosome preparation by “conventional” techniques.

Preparation Method description Reference

Thin layer evaporation (TLE) The formation of a surfactant:lipid film by the evaporation of an organic solution of surfactants:lipids. This film is then [61]
hydrated with aqueous solutions to obtain multilamellar vesicles which are further treated for size/lamellae reduction.
Disadvantage: use of organic solvent.
Advantage: easy technique for bench researches.
Melted amphiphile injection The injection of melted lipids:surfactants into a highly stirred heated aqueous phase. [62]
Disadvantage: no applicability for thermosensitive drugs.
Advantage: no organic solvents, which are expensive, difficult to remove completely and hazardous.
Solid amphiphiles/hot water The addition of the warmed aqueous phase to a mixture of the solid lipids:surfactants. [63]
Disadvantage: no applicability for thermosensitive drugs.
Advantage: no organic solvents, which are expensive, difficult to remove completely and hazardous.
Micellar solution/enzymes Niosomes may also be formed from a mixed micellar solution by the use of enzymes. [64]
Disadvantage: enzymatic degradation of active substances.
Advantage: no organic solvents, which are expensive, difficult to remove completely and hazardous.
Ether injection In this method, the surfactans and drug are dissolved in diethyl ether and injected slowly into an aqueous phase, which is [65]
heated above the boiling point of the organic solvent. This produces large unilamellar vesicles, which are further treated for
size/lamellae reduction
Disadvantage: no applicability for thermosensitive drugs.
Advantage: easy technique for bench researches.
Trans membrane pH gradient method After film formation (TLE) a solution of an acidic compound (generally citric acid) is added and mechanically stirred Freeze- [11]
thawing cycles are then carried out on the obtained product after which an aqueous solution of the drug is added and the
mixture is vortexed. pH is then finally adjusted to 7–7.2 by addition of a buffer solution.
Disadvantage: use of organic solvent.
Advantage: high drug entrapment efficiency.
Reversed phase evaporation The surfactants are dissolved in a mixture of ether and chloroform and an aqueous phase containing the drug is added. The [11]
resulting two-phase system is then homogenized and the organic phase evaporated under reduced pressure to form
niosomes dispersed in the aqueous phase.
Disadvantage: use of organic solvent
Advantage: high drug entrapment efficiency
Bubbling of nitrogen Nitrogen is passed through a sample of homogenized surfactants leading to the formation of large unilamellar vesicles, which [66]
are further treated for size/lamellae reduction.
Advantage: no organic solvent.
Microfluidization A solution of surfactants and drug is pumped under pressure from a reservoir through an interaction chamber packed in ice at [67]
a rate of 100 ml/min. From the interaction chamber, the solution is passed through a cooling loop to remove the heat
produced during micro fluidization.
Disadvantage: no applicability for thermosensitive drugs.
Advantage: no organic solvents, which are expensive, difficult to remove completely and hazardous.
C. Marianecci et al. / Advances in Colloid and Interface Science 205 (2014) 187–206 193

Permeation under
Pressurized vessel Organic phase
applied pressure
Dispersed phase

Tangential flow Membrane


N2 bottle

Niosome

Pump
Magnetic
stirrer

Aqueous phase

Fig. 6. Schematic drawing of the membrane experimental set-up for niosome preparation.

two miscible fluids (i.e., alcohol and water) in microchannels. Different removed. Although it may be argued that the use of systems in which
parameters might affect assembly of niosomes, such as (1) conditions the drug is partially encapsulated in niosomes may eventually yield
for the microfluidic mixing, (2) chemical structures of the surfactant systems with a beneficial biphasic biodistribution profile. This drug
used (i.e., Sp 20, Sp 60, and Sp 80), and (3) device materials for the delivery system could lead to an initial burst effect to initiate therapy
microchannel fabrication [60]. followed by a sustained maintenance dose. The methods used for the
removal of unentrapped material include different techniques:

4.2. Vesicle purification 4.2.1. Dialysis


Dialysis is based on diffusion and osmosis of solutes and fluid across
The hydration of niosome mixtures seldom leads to complete drug a semi-permeable membrane. Several authors used this technique
encapsulation, regardless of the drug loading optimization steps under- to purify their vesicle dispersion from unentrapped materials: the
taken. It is thus often a requirement that unencapsulated drug must be prepared niosomes are filled into dialysis bags and the free drug is
dialyzed against buffer saline [30,68,69].

4.2.2. Gel filtration


In order to separate loaded NSVs from unentrapped substances,
Phosphate Phosphate purified vesicle dispersion can be obtained by means of gel-filtration
Buffer Buffer on Sephadex G75 or G50 or Sephadex G25 [70–76].
Gel-filtration chromatography is a popular and versatile technique
that allows the effective separation of free molecules in high yield.

4.2.3. Centrifugation/ultracentrifugation
To purify niosomes, the centrifugation and ultracentrifugation sepa-
ration was reported [77].
In particular the centrifugation process was carried out to remove
the unentrapped genetic material from niosomes, following the gradi-
ent density centrifugation approach [78–81].
Some authors combined the previously mentioned centrifugation
processes with Sephadex chromatography mini-columns (Minicolumn
centrifugation method) [80,81]. This procedure compared to other
purification methods is applicable to a variety of solutes and 92 to
100% recovery can be achieved for niosomes with no dilution of the
niosomal preparation.
Since all these methods show, at the same time, advantages and
disadvantages, consequently the choice of the method must take into
account different factors. For instance, for industrial purposes it may
Fig. 7. Schematics of niosome self-assembly by microfluidic hydrodynamic focusing. A be more appropriate to concentrate efforts and resources on the
central stream containing the surfactant mixture in isopropyl alcohol is focused by
adjacent streams of phosphate buffer in a microfluidic format. FRR = flow rate; QB =
achievement of high levels of drug loading, thus avoiding separation
buffer volumetric flow rates; QS = alcohol volumetric flow rate; Wf = width of the steps or to consider systems in which the unentrapped drug may be
focusing stream; τmix = diffusive mixing time. useful as a specific priming dose.
194 C. Marianecci et al. / Advances in Colloid and Interface Science 205 (2014) 187–206

4.3. Vesicle characterization 4.3.3. Bilayer characterization


Niosomes, as liposomes, can be morphologically classified according
A rational characterization of nanosystems is needed for the control to the number of membrane bilayers (lamellae) in uni- and multi-
of the quality of the product, which is a prerequisite for their develop- lamellar vesicles. Uni-lamellar vesicles are characterized by the pres-
ment and clinical applications. Having direct impact on the stability ence of one surfactant bilayer while multi-lamellar vesicles by several
and critical importance on their in vivo performance, niosomes param- concentric surfactant bilayers in an onion-like skin arrangement. The
eters such as morphology, size, polydispersity index, number of lamel- number of lamellae can be determined by AFM, NMR and small angle
lae, ζ-potential, bilayer fluidity, composition, encapsulation efficiency, X-ray scattering (SAXS). The latter method, together with the in situ
carrier-bioactive interaction and chemical stability must be evaluated energy-dispersive X-ray diffraction (EDXD), has been also used to char-
(Table 2). acterize the bilayer thickness [84,85,92–95].
Another feature of the niosomes is the fluidity of their membrane,
4.3.1. Vesicle size which allows membrane deformation without disrupting bilayer integ-
Determination of nanocarrier size distribution is a fundamental rity (vesicle stability) and drug-release ability.
parameter in the determination of physical properties and stability of These parameters can be measured by means of the mobility of fluo-
bioactive delivery formulations. Size distribution, along with composi- rescence probes (e.g. 1,6-diphenyl-1,3,5-hexatrine, DPH, and pyrene) as
tion, defines plasma pharmacokinetics, biodistribution, and stability of a function of temperature and/or time [85,96–98].
nanocarriers and their associated/entrapped substances in plasma and The microviscosity of niosomal membrane can also be determined
other organs [82]. by fluorescence polarization in order to study packing structure of the
Niosome diameters can be determined using microscopy or dynamic vesicular membrane [58].
light scattering (DLS or photon correlation microscopy, PCS).
DLS provides simultaneously cumulative information of the average 4.3.4. Vesicle stability
particle and of the particle size distribution. One problem with DLS is Vesicle stability is a complex issue and involves chemical stability,
that it does not provide information on the shape of the nanosystem physical stability and biological stability, which are all inter-related.
and it assumes any aggregation of more than one particle as one single The evaluation of these parameters is fundamental to determine the
unit. The microscopic approach is commonly used to characterize the potential in vitro/in vivo applications in nanomedicine. Biological stabil-
structure/morphology/geometry of the nanocarriers. ity, however, depends on the presence of agents that interact with the
Several electron microscopy techniques can be employed for vesicular structure after administration and it therefore depends also
nanocarrier characterization. These include: scanning electron micros- on the administration route. Generally, stability is determined by
copy (SEM), preferentially for solid products; transmission electron means of size and ζ-potential variations (DLS, Turbiscan Lab® Expert
microscope (TEM), for liquid state sample; negative-staining transmis- or microscopy techniques) or by evaluation of the release rate of differ-
sion electron microscope (NS-TEM); freeze fracture transmission elec- ent probes as a function of time and/or temperature, in absence or in
tron microscopy (FF-TEM); cryo-transmission electron microscopy presence of biological fluids [99].
(cryo-TEM), atomic force microscopy (AFM) and scanning tunneling
microscope (STM), used in 1982 by Binnig and coworkers, for the 4.3.5. Entrapment efficiency
study of micro- and nano-scale structures. Furthermore, STM in partic- The entrapment efficiency (EE) of vesicular systems can be defined
ular is very useful in determining the bilayer thickness of liposomes, as the amount of active substances loaded within the niosomal structure
nano-liposomes, and niosomes by its analytical ability in the vertical and can be expressed as:
axis [30,31,70,71,83–88].
It must be pointed out that microscopy techniques can give artifacts amount entrapped
EE ¼  100 ð2Þ
and hence it can be useful to combine different techniques to obtain total amount
more reliable results.
where “total amount” is the total amount of drug used in the prepa-
4.3.2. ζ-potential and surface properties ration. The entrapment efficiency can be determined by spectropho-
ζ-potential value has critical importance on vesicle stability and tometry or, in the case of genetic materials (e.g. luciferase plasmid),
in vivo fate. Several authors confirmed the importance of ζ-potential by gel electrophoresis followed by UV densitometry. In addition, the
measurements to assess vesicle formation, to study drug/vesicle inter- entrapment efficiency can be fluorometrically evaluated using a hydro-
action and formulation stability [70,71,89]. philic fluorescent marker (e.g. calcein) and then calculated according to
ζ-potential is also a critical parameter to be taken carefully into the following equations
account in the formation of aggregates, from charged niosomes and
oppositely charged polyions, when these vesicles are proposed as drug Moleprobe entrapped
EE ¼  100 ð3Þ
carriers [90,91]. Molesurfactant

Table 2
Different approaches to niosome characterization.

Niosome parameter/property Applied technique Reference

Size DLS/PCS; SEM (preferentially for solid products); [30,31,70,71,82–88]


TEM (liquid state sample); NS-TEM; FF-TEM; cryo-TEM; AFM; STM
ζ-potential Electrophoretic mobility. [70,71,89–91]
Bilayer characterization Fluorescence polarization. [59,71,84,85,92–95]
Fluorescence anisotropy.
AFM; NMR; SAXS; EDXD.
Stability DLS; Turbiscan; microscopy techniques. [154,99]
Release studies.
Entrapment efficiency UV/VIS; HPLC; fluorescence. [30,37,68]
For genetic material: gel electrophoresis followed by UV densitometry.
pH-sensitivity assessment Fluorescence spectroscopy; NRET; SAXS [91,102–104]
C. Marianecci et al. / Advances in Colloid and Interface Science 205 (2014) 187–206 195

lin −ltx  r  100 and targeting vary by cell or tissue type, so the behavior of niosomes
EE ¼  100 ð4Þ
ltotal −ltx  r in vitro/in vivo may not be predictive of another.
An ever increasing number of publications on potential and practical
niosome applications in nanomedicine appeared in the scientific litera-
ture during the last years and most of them take into account the impor-
where Itotal and Iin are the fluorescence intensity before (Itotal) and after tance of nanovector characterization.
(Iin) addition of cobalt chloride (CoCl2) respectively, Itx is the fluores- To highlight the state of the art of niosome pharmacological and
cence intensity after addition of CoCl2 and a detergent such as diagnostic applications, we attempt to summarize in this section cur-
n-propanol or Triton X-100, r is the volume correction factor (~1.04). rently available information on niosomal carriers that are endowed
The second equation does not require vesicle purification [30,37,68,100]. with desiderable properties.

4.3.6. pH-sensitivity assessment 5.1. Dermal and transdermal delivery


Stimuli-sensitive NSVs, which release their cargo in response to
external stimuli, represent interesting alternatives for therapies direct- Dermal drug delivery is the topical application of drugs to the skin in
ed towards solid tumors and other spatially well defined targets. The the treatment of skin diseases.
gradual decrease in pH, experienced by niosomes that are internalized This approach has the advantage that high drug concentrations can
via endocytosis, is actually a very useful intrinsic stimulus and several be localized at the site of action, minimizing systemic absorption, thus
pH-sensitive niosome formulations, based on this strategy, have been reducing systemic side effects. On the other side, transdermal drug
developed and biologically evaluated [91,101,102]. delivery uses the skin as an alternative route for the drug towards the
The steady-state fluorescence spectroscopy was also used to charac- systemic circulation. This drug delivery route shows several advantages
terize the pH-sensitivity of the surfactant vesicles. The fluorescent over the conventional oral and parental routes such as: avoidance of the
probes chosen for such experiments were hydroxypyrene-1,3,6- risk and inconvenience of intravenous therapy (non invasive), avoid-
trisulfonic acid (HPTS) and Nile Red. The maintenance of pH-sensitivity ance of first pass hepatic metabolism (avoiding the deactivation by
in serum must be appropriately verified by monitoring fluorescent digestive and liver enzymes), which leads to an increase of drug bio-
probe release at different pH in the presence of serum. The pH effect availability and efficacy, no gastrointestinal degradation (pH, enzymatic
on vesicle bilayer can be also evaluated by SAXS. This technique is able activity, drug interaction with food, beverages and other orally adminis-
to evaluate bilayer thickness changes in different pH environments tered drugs), alternative to oral administration when such route is
[91,103]. unsuitable (e.g., vomiting and diarrhea). However, transdermal drug
An assay, performed at different pH, for vesicle-vesicle fusion evalu- delivery shows the major disadvantage of a low penetration rate
ation, is based on the non radioactive resonance energy transfer (NRET) through the skin. Actually, only limited number of drugs can be formu-
between two fluorescent lipid analogs N-(7-nitrobenz-2-oxa-1,3- lated as transdermal delivery systems due to the functions of the stra-
diazol-4-yl)-1,2-dihexadecanoyl-snglycero-3-phosphoethanolamine tum corneum (SC), which provides the main barrier to permeation.
(NBD-PE) (energy donor), and N-(lissamine rhodamine B sulfonyl)- Although a number of penetration enhancement techniques like
phosphatidylethanolamine (N-Rh-PE) (energy acceptor). This tech- iontophorosis, ultrasound (phonophoresis and sonophoresis),
nique provides a means by which lipid mixing during vesicle fusion magnetophoresis, electroporation, laser radiation and photomechanical
can be followed since fusion of the vesicles will result in intermixing waves have been used, the number of drugs that can be administered by
of the membrane lipids and probe dilution [104]. transdermal route is still limited. During the last decade, vesicle applica-
tion in drug delivery through the skin has undergone intense investiga-
5. Niosomes in drug delivery and targeting tions and it seems to be promising [105–109] (Table 3). Both a
permeation enhancer effect and direct vesicle fusion with the SC may
Niosomal carriers are suitable for the delivery of numerous pharma- contribute to the enhanced permeation of drugs loaded in niosomes.
cological and diagnostic agents. According to this statement it must be NSVs are becoming popular in the field of topical drug delivery due to
pointed out that an emerging paradigm for the design of effective their outstanding characteristics and properties induced by their pres-
nanocarriers is the modification of physico-chemical parameters to ence in the formulation such as enhanced drug penetration, sustained
influence the drug delivery and targeting. These factors are often drug release, increased drug stability and ability to carry both hydrophil-
difficult to vary independently, so the contribution of each is difficult ic and lipophilic drugs [110]. Fang et al. showed that enhancing effect for
to generalize. Furthermore, processes involved in efficient drug delivery skin permeation of enoxacin for niosomes was greater than that of

Table 3
Dermal and transdermal application of niosomes.

Compositions Drug Experimental model Results Reference

Span40/Chol, Span60/Chol, Enoxacin In vitro diffusion: Franz cells — nude mouse skin. Niosome fusion at the interface of the stratum [111]
Span60/DCP/Chol corneum: high local drug concentration.
Span40/Chol/DCP, Span60/Chol/DCP, Tretinoin In vitro permeation: Franz cells — silicone membrane High local drug concentration. High trenitoin [74,75]
Brij30/Chol/DCP, (TRA) (Perthese®). permeation Best formulation: TritonCG 110-
TritonCG110/Chol/DCP Niosomes
Oramix®CG110 or In vitro permeation: Franz cells — new-born pig skin. Drug delivery modulated by varying the structure
Oramix®NS10/Chol/DCP or SA and/or bilayer composition: highly hydrophilic
surfactants (HLB = 16) improve TRA diffusion
through pig skin; Brij® 30 or Oramix NS 10 (HLB 9 and
11, respectively) enhance TRA cutaneous retention
Tw20/Chol, Tw20/Chol/Chems, Ibuprofen In vitro permeation: Gummer cells — rat skin. The more lipophilic the surfactant, the greater its [113]
Sp60/Chol, Sp60/Chol/Chems ability to penetrate into the skin
Span60/Chol in hydrogel Meloxicam Carrageenan induced rat paw edema method. Marked increase in the percentage inhibition of edema [124]
in animals treated with meloxicam vesicular gel.
Span40/Chol niosomal gel Lopinavir Ex vivo skin permeation studies; histopathological Better safety, bioavailability and deposition in [123]
studies; in vivo bioavailability study in male Wistar comparison to ethosomal gel
rats.
196 C. Marianecci et al. / Advances in Colloid and Interface Science 205 (2014) 187–206

liposomes composed of dimyristoyl phosphatidylcholine. Manconi et al. Mucoadhesive timolol maleate NSVs coated with chitosan or
showed that higher tretinoin stability was obtained when it was incor- carbopol have been used to extend optimal ocular pharmacodynamics
porated in niosomes rather than in liposomes [111,112]. over a prolonged period, and they showed only limited systemic
Recently pH-sensitive NSVs obtained with Tw20 or Sp 60 mixed absorption and side effects [134,135].
with CHOL and cholesteryl hemisuccinate (CHEMS), a derivative of Niosomal formulations, prepared using various surfactants (Tw20,
cholesterol as a pH-sensitive molecule, were proposed for topical Tw60, Tw80 or Brij 35) showed a slower in vitro release of gentamicin,
delivery of ibuprofen. Only niosomes with Sp 60 and CHEMS showed a when compared to the drug solution or promote absorption of
significant increase of in vitro skin permeation of the drug [113]. cyclopentolate by preferentially modifying the permeability character-
Other drugs loaded into niosome for skin administration were: istics of the conjunctival and scleral membranes [136,137].
gallidermin; terbinafine; curcumin; vinpocetine; ellagic acid and am- Recently, Sp 60-based oval giant niosomes were reported to be
monium glycyrrhizinate [114–120]. capable of controlling naltrexone hydrochloride permeation and
An interesting study reports a comparison between lopinavir enhance its corneal permeability and were found practically non irritant
niosomal gel and ethosomal gel formulations. The niosomal formulation [138].
containing lopinavir, Sp 40, and CHOL possessed a high entrapment In the study NSVs and discomes for ocular delivery of naltrexone
efficiency for the drug with a minimum mean vesicular diameter. hydrochloride (NTX) were evaluated for their spreading, rheological
Ex vivo skin permeation studies of lopinavir, as well as those of the properties and their ability to prevent NTX autoxidation in aqueous
fluorescent probe coumarin, revealed a better deposition of ethosomal solutions. The prepared niosome formulation was significantly more
carriers but a better release with niosomal carriers. Histopathological viscous than the aqueous solution. The lipid content, size and composi-
studies indicated the better safety profile of niosomes over ethosomes. tion of NSVs were the main factors affecting the viscosity of the
In vivo bioavailability study in male wistar rats showed a significantly niosomal dispersions. Exposure of NTX solution to artificial daylight
higher extent of absorption of lopinavir via transdermally applied illumination can produce extensive degradation of NTX due to oxida-
niosomal gel when compared with its oral suspension. Taken together, tion, while the prepared formulations were able to significantly protect
these findings suggested that niosomal gel holds a great potential of the encapsulated NTX from the photo-induced oxidation when
being used as novel, nanosized drug delivery vehicle for transdermal compared with free NTX solutions. The investigated niosomes showed
lopinavir delivery [121]. a potential ocular delivery for NTX.
To enhance transdermal adsorption of drugs, elastic NSVs, niosome Furthermore, NTX loaded-niosomes were proposed as a promising
loaded hydrogel systems [122–124] and proniosomal gel were also treatment for corneal disorders linked to diabetes mellitus (diabetic
proposed [125–130]. Topical immunization is novel and needle free keratopathy) [139].
strategy involving vaccine delivery through topical application of anti- Niosomal formulations showed a fairly high retention of acetazol-
gen and adjuvants directly or via a suitable carrier system on intact amide inside the vesicles (75%) at a refrigerated temperature for up to
skin. Anatomy and physiology of skin attracts scientists in developing three months. Each of the tested acetazolamide niosomes produced
topical carrier systems for enhanced delivery of bioactive substances significantly less intraocular pressure than the free drug and basic
and niosomes have been exploited for topical immunization [131,132]. niosome solution [140]. Studies of aqueous humor pharmacokinetics
The application of niosomes as dermal/transdermal delivery systems and acetazolamide niosomes revealed that the Cmax of the drug from
is receiving an increasing attention and they are becoming popular in the niosome formulation was double than that of the drug suspension,
the field of topical delivery due to their outstanding characteristics showing a significant broadening of the peak from 80 to 120 min. The
like enhancing the penetration of drugs and providing a sustained pat- concentration remained over 13 μg throughout this period [141].
tern of drug release [133]. It was demonstrated that deformable elastic liposomes can enter the
corneal structure that is similar to the SC [142] and that elastic niosomes
5.2. Ocular delivery show advantages over liposomes and over conventional NSVs as well. In
that, the former can reach even the posterior eye segment due to its
There are many anatomical and physiological barriers to overcome elasticity and nanosize. This elasticity is attributed to the use of edge
in topical ocular drug delivery and a strategy to cross them implies the activators (EAs). EAs are the surfactant molecules that provide flexibility
use of bioadhesive carriers (Table 4). to the lipid bilayer membranes of these vesicles. These surfactant

Table 4
Niosomes in ocular delivery.

Compositions Drug Experimental model Results Reference

Chitosan or carbopol coated niosomes Timolol maleate In vitro release: dialysis bag; in vivo Significant decrease in IOP [134,135]
(Span 60:Chol 1:1) pharmacodynamic studies (intra ocular
pressure, IOP) on adult male normotensive
rabbits
Tween 60 or Tween 80 or Brij 35/Chol/DCP Gentamicin In vitro release: dialysis bag; in vivo evaluation Niosomes exhibit an alkyl chain length- [136]
of potential ocular irritancy and/or damaging dependent release, the higher the chain
effects on albino rabbits. length, the lower the release rate. DCP stabi-
lizes the niosomal membrane and delays the
drug release
Tween 20/Chol Cyclopentolate In vitro permeation — rabbit corneas; in vivo In vitro enhanced transcorneal permeation; [135]
evaluation of mydriatic activity in rabbits in vivo improved drug ocular bioavailability
Niosomes: Span 60/DCP or Sodium Cholate Naltrexone hydrochloride Evaluations on surface tension, contact angle, Enhanced corneal uptake of the hydrophilic [137,138]
Discosomes: Span 60/Solulan C24 spreading coefficient, viscosity and effect of drug (NTX). Enhanced NTX protection from
niosomal encapsulation on NTX oxidation. photo-oxidation compared with conventional
NTX aqueous solutions.
Span 40 or Span 60, or Span 80/edge Fluconazole In vitro genotoxicity and cytotoxicity; ex vivo Elastic niosomes safety in terms of [144]
activators (EAs): sodium taurocholate or corneal permeability studies (porcine cornea); genotoxicity, cytotoxicity, acute dermal/eye
sodium cholate or sodium deoxycholate, in vivo ocular irritation studies in rabbits. irritation/corrosion, and chronic eye irritation/
or Tween 80 corrosion test. Enhanced drug corneal
permeability
C. Marianecci et al. / Advances in Colloid and Interface Science 205 (2014) 187–206 197

molecules destabilize the lipid bilayers and, in turn, increase the the oral bioavailability of griseofulvin in albino rats after a single oral
deformability of the vesicles [143]. The surfactant present in these dose [146].
vesicles induces pores in lipid structures, such as membranes, and also Recently, NSVs from Sp 40, Sp 60, and CHOL were proposed as
leads to a solubilization (lysis) at the higher concentration range. potential oral delivery systems for the effective delivery of ganciclovir.
In the study of Kaur et al. [144] Sp-based elastic (spanlastic vesicles) In vivo study on rats revealed a five-time increase in bioavailability of
novel vesicular systems were developed and proposed as carrier the drug after oral administration with respect to tablets [147].
systems for fluconazole, which is a hydrophilic drug with a short Metformin is an oral hypoglycemic which is widely used as a first
ocular elimination half-life and low log P value (log P = 0.25). A better line treatment of type II diabetes, but at a very high dose 2–3 times a
permeation was achieved by these systems and can be explained ac- day; moreover it suffers from a number of side effects such as lactic
cording to two transport mechanisms. First, the elastic vesicles interact acidosis, gastric discomfort, chest pain, allergic reactions. To decrease
with the epithelial cell membrane and act as penetration enhancers, metformin side effects, and dose frequency, niosomal formulations
and subsequently modify the intercellular lipid lamellae (mechanism obtained with Sp 60 and Sp 40 and CHOL in equimolar ratio were pro-
1). Second, the elastic vesicles can act as drug-carrier systems, whereby posed. The obtained vesicles showed a high entrapment efficiency and
intact vesicles carrying the drug pass through the intercellular spaces a sustained release of the drug over extended period of time; thus signif-
and reach the biological membrane (mechanism 2). icantly improving the performances of the drug [148].
The developed spanlastic systems were found to be safe in terms of NSVs were also used for oxcarbazepine loading with the aim of
genotoxicity, cytotoxicity, acute dermal/eye irritation/corrosion, and improving its anticonvulsant activity. The in vivo study indicated how
chronic eye irritation/corrosion tests. the dispersion of drug-loaded vesicles, prepared by the thin film hydra-
tion method, showed an improved anticonvulsant activity in albino
5.3. Oral delivery wistar rats together with a significant increase in the mean residence
time, thus reflecting sustained release properties [149].
Oral route is the preferred route of administration for most drugs; Lornoxicam loaded niosomes were prepared by thin film hydration
however, the majority of the newly discovered and existing drugs method with different Sp 60:CHOL ratios and the obtained formulations
administered by oral route frequently must face bioavailability prob- were evaluated by and for their entrapment efficiency. In vitro release,
lems due to several reasons such as poor solubility and low dissolution kinetic data analysis and stability studies suggested that multilamellar
rate, unpredictable absorption, inter and intra subject variability niosomal formulation could provide consistent and prolonged release
and lack of dose proportionality. Several strategies have been adopted of lornoxicam from different niosomal formulations. Such sustained
for the improvement of dissolution behavior of insoluble drugs; activity of the entrapped drug may lead to a reduction of the side effects
e.g., complexation, drug derivatization, solid state manipulation, inclu- associated with frequent administration and correspondingly potenti-
sion of surfactants, increase of the surface area by micronization or ate its overall the therapeutic effects [150]. An interesting study focused
nanonization, spray drying and microencapsulation (Table 5). on the evaluation of the stability of NSVs composed by Tw20 and CHOL,
NSVs are also capable to enhance drug stability in the gastrointesti- in the artificial gastrointestinal fluid was performed by Di Marzio et al.
nal environment; in this sense it was reported that the in vitro gastroin- Tw20 is a non-ionic surfactant, the stability and relative non-toxicity
testinal stability of paclitaxel was well preserved with Sp 40 NSVs [145]; of which, allows it to be used as excipient for oral administration.
moreover vesicles from Sp 20, Sp 40, and Sp 60 significantly improved From in vitro simulations, it was evidenced that Tw20 vesicles show

Table 5
Niosome in oral delivery.

Compositions Drug Experimental model Results Reference

Tweens or Spans or Brijs/Chol and DCP Paclitaxel In vitro PCT release; stability studies in GI en- Fast drug release from Tween niosomes (more flexible [145]
(PCT) zymes. structure). Best PCT protection against GI enzymes with
Span 40 niosomes
Span 40 or Span 60/Chol Ganciclovir In vitro drug release, release kinetic study, and In vivo study on rats reveals five-time increment in bio- [147]
in vivo performance availability of Ganciclovir after oral administration of op-
timized formulation (Span60:Chol 3:2) as compared with
tablet.
Spans or Tweens or Brijs/Chol/DCP Metformin In vitro release studies with USP dissolution Span 60 having longer chain length provide more stable [148]
apparatus. vesicles, higher entrapment and delayed release.
Spans/Chol Oxcarbazepine Pharmacokinetic studies in Albino Wistar rats Increased elimination half life and AUC when compared [149]
with free drug
Span 60/Chol Lornoxicam In vitro drug release studies. Consistent and prolonged release of lornoxicam. [150]
Span 60/Chol/coating agent (maltodextrin Glipizide In-vitro release — dialysis. Consistent and prolonged release with maltodextrin- [152]
or sorbitol or mannitol): proniosomes based proniosome
Span 60/Chol/DCP: proniosomes Celecoxib Dissolution studies; pharmacokinetic Slow drug release; Significantly higher values and [153]
evaluations capsules in comparison to significantly lower values for absorption rate constant
Celebrex®, Pfizer, on six healthy male. (Ka) of the proniosomal system compared with
Celebrex® capsule.
Span 60/Chol/Maltodextrin:proniosomes Valsartan In vitro dissolution studies in SGF and SIF; ex- Drug improved dissolution rate; enhanced drug [155]
vivo permeation studies on male Wistar rats' absorption across gastrointestinal membrane.
ileum.
Monopalmitoylglycerol/Chol/DCP/bile salts: Influenza In vivo biodistribution and vaccine efficacy Increase uptake within the Peyer's patches and reduction [157]
bilosomes vaccine studies in viral cell load in an influenza challenge study.
Tw20/Chol Tw20/Chol/Chems = Stability evaluation in SIF and SGF. Assess- Good stability in SIF and SGF; good mucoadhesive prop- [151]
ment of mucoadhesive properties. erties in SIF rather than in SGF
Span 60/Chol/stearylamine niosomes, Plasmid DNA In vitro ligand-specific activity by concanava- Measurable humoral and cellular immune response upon [158]
emulsificated with aqueous solution of encoding for lin A agglutination assay. Stability studies in oral administration.
plasmid DNA, coated with o-palmitoyl HbsAg. SGF and SIF. Biodistribution studies on BALB/c
mannan. mice
198 C. Marianecci et al. / Advances in Colloid and Interface Science 205 (2014) 187–206

good stability in simulated gastric and intestinal fluid, also in the pres- deoxycholate at different blend ratios. The optimized bilosome formula-
ence of digestive enzymes and bile salts, as evidenced by the release tion was identified and the potential of this formulation as an oral
of a fluorescent probe which is most likely related to small membrane vaccine delivery system was assessed by biodistribution and vaccine
damages rather than to a disintegration of the vesicles. Furthermore, efficacy studies. Obtained results showed that the larger bilosomes
these vesicles were able to adhere to mucin in the intestinal medium vesicles increased uptake within the Peyer's patches and promoted a
more than in gastric one [151]. reduction in viral cell load in an influenza challenge study [157].
Approaches to stabilize niosomal drug delivery systems without Mannosylated niosomes were used as oral DNA vaccine carriers for
affecting their properties led to the development of the promising the induction of humoral, cellular and mucosal immunity. NSVs, com-
drug carriers called “Proniosomes”. Proniosomes are a dry formulation posed of Sp 60 with the addition of CHOL and SA as constitutive lipids,
obtained from a suitable carrier coated with non-ionic surfactants were prepared by reverse phase evaporation and were coated with a
which can be converted into NSVs immediately before use by hydration. modified polysaccharide o-palmitoyl mannan (OPM) in order to protect
These proniosome-derived niosomes are as good as or even better than them from bile salt-caused dissolution and enzymatic degradation in
conventional niosomes. Glipizide loaded sorbitol, maltodextrin and the gastrointestinal tract and to enhance their affinity towards the anti-
mannitol based proniosomes were prepared by a slurry method with gen presenting cells of Peyer's patches. OPM coated niosomes produced
different surfactant:CHOL ratios [152]. humoral (both systemic and mucosal) and cellular immune response
Another proniosome formulation (Sp 60:CHOL:DCP, 1:1:0.1) was upon oral administration. This study signifies the potential of OPM
studied to improve the extent of celecoxib absorption, compared to coated niosomes as DNA vaccine carriers and adjuvants for effective
that of a conventional marketed celecoxib capsule. Experiments, carried oral immunization [158].
out on human volunteers, showed that the mean relative oral bioavail-
ability of the proniosomal formulation increased with respect to that of 5.4. Pulmonary delivery
the conventional capsule [153]. Similar results were reported by Xiao
et al. who studied the effect of oral proliposome on silymarin bioavail- In inflammatory diseases, infections or cancer of the respiratory
ability [154]. tract, the therapeutic value of pulmonary administration may exceed
The oral administration of valsartan proniosomes was also studied to that of an oral or parenteral administration (Table 6) and in diseases
overcome its low absolute bioavailability (25%) due to the poor aqueous characterized by bronchial mucus hypersecretion, lipophilic substances,
solubility, as well as the drug side effects in the gastrointestinal tract and such as corticosteroids, can be remarkably impeded in reaching their
the fact that two third of the dose is excreted by renal and biliary trans- receptors, which are localized within the cytoplasm of bronchial epithe-
port. Jukanti et al. reported the formulation and characterization of lial cells. Surfactant vesicles were proposed to circumvent this prob-
valsartan loaded maltodextrin based proniosomal powders. Ex-vivo lem [73]. Niosomal delivery of beclomethasone dipropionate (BDP) in
studies, carried out on rat intestine to assess the permeation of valsartan asthma and chronic obstructive pulmonary disease was evaluated by
from proniosomes showed a significant permeation enhancement different research groups.
across rat intestine thus suggesting the potential of proniosome carriers Terzano et al. [73] and later Marianecci et al. [159] carried out formu-
for improved oral delivery of valsartan [155]. lation, physical–chemical and in vitro cell characterization studies on
NSVs have also been tested as stabilizers for therapeutic proteins BDP loaded Tw20 vesicles in order to evaluate the possible advantages
and peptides in the gastrointestinal tract and for a possible improve- of this new type of NSV system that can lead to an improved mucus per-
ment of their permeation through the intestinal mucosa. In an in vitro meation. Actually, such delivery system should enhance permeation
study by Yoshida et al. oral delivery of a vasopressin derivative through mucosal barriers because of the presence of vesicles formed
entrapped in niosomes showed that vesicle inclusion significantly with a remarkably hydrophilic non-ionic surfactant usually considered
increased the stability of the peptide, in an intestinal loop model [156]. as unsuitable for the formation of vesicular structures because of its
Oral vaccines offer significant benefits due to the ease of administra- high HLB value (HLB = 16.7). From the obtained results, it was
tion, better patient compliance and non-invasive, needle-free adminis- highlighted that the vesicular dispersions show rheological characteris-
tration. However, this route is marred by the harsh environment tics and nebulization efficiency (i.e. aerodynamic diameter and drug
which is detrimental to many vaccine formats. To overcome such prob- mass output) comparable with that of a commercial product. Further-
lems, bilosomes have been considered; these are bilayer vesicles more, the novel preparation, ensuring a better penetration through
constructed from non-ionic surfactants combined with the inclusion the mucus layer, can provide an efficient system for the treatment of
of bile salts which can stabilize the vesicles in the gastrointestinal tract obstructive pulmonary diseases. The aim of the second part of the
by preventing membrane destabilization. Bilosomes were actually same study, carried out by Marianecci et al., was the evaluation of the
constructed from monopalmitoylglycerol, CHOL, DCP and sodium interaction between the innovative NSVs and human lung fibroblast

Table 6
Niosomes in lung delivery.

Compositions Drug Experimental model Results Reference

Tween 20/Chol Beclomethasone In vitro permeation experiments (mucin solution 0.1% Increased permeation rate through the model [73,159]
dipropionate w/v). Aerosolized droplet size studies. Cellular tolerability mucosal barrier. Good in vitro tolerability on HLFs.
and cytotoxic on primary human lung fibroblasts (HLFs). Improved drug anti- inflammatory activity in HLFs.
Evaluation of antiinflammatory effect on HLFs.
Span 60/Chol/sucrose: Beclomethasone Nebulization by Pari LC Sprint (Air-jet), Aeroneb Pro High drug output and fine particle fraction (FPF) by all [56]
proniosomes dipropionate (Actively vibrating-mesh) nebulizer and Omron MicroAir devices
NEU22: aerosol mass output gravimetrical determination
and aerosol droplet size analysis.
Tween80/Chol niosomes Amphotericin B In vitro antifungal and antileishmanial activity of AMB- Significant reduction in fungal lung burdens in a rat [169]
encapsulating niosomes; in vitro AMB delivery studies. In vivo real time model of invasive pulmonary aspergillosis and in a
hydroxypropyl-γ- delivery studies; in vivo antileishmanial and anti asper- significant suppression of Leishmania donovani liver
cyclodextrin/AMB gillosis activity of AMB-niosomes parasite burden
Span 60 and Tween 60/Chol Ciprofloxacin HCl MIC determination against some pulmonary pathogens. In vitro safety and efficacy of Ciprofloxacin loaded [170]
MTT assay in human lung carcinoma cell line. niosomes
C. Marianecci et al. / Advances in Colloid and Interface Science 205 (2014) 187–206 199

(HLF) cells, the carrier tolerability, the vesicle localization within the In order to develop a niosome-encapsulated ciprofloxacin HCl
cells and the amount of BDP actually internalized by the cells. The (CPFX) formulation for pulmonary delivery, the feasibility of CPFX en-
study confirmed that BDP-loaded NSVs were able to improve the drug capsulation in niosomes, their stability and nebulization process
anti-inflammatory activity in HLFs due to the carrier ability to increase effectiveness was evaluated by Moazeni et al. [170]. Various combina-
the intracellular delivery of BDP and hence to determine a great amount tions of non-ionic surfactants with cholesterol were used to prepare
of drug interacting with its cytoplasm receptor. The overall conclusion the formulations. The in vitro deposition data of the niosomal formula-
of this investigation was that these innovative NSVs could be considered tions were examined using an andersen cascade impactor. Formulations
as effective and safe carriers for the BDP pulmonary delivery, thus suit- composed of Sp 60 and Tw60 in combination with 40 mol% of CHOL
able for the treatment of pulmonary inflammatory diseases. exhibited high encapsulation efficacy and stability and also had fine
Proniosomes were investigated also for pulmonary delivery of BDP. particle fraction and high nebulization efficiency. Minimal inhibitory
Hence, proniosomes represent an approach to formulate stable pow- concentration of the niosomal CPFX against some pulmonary pathogens
dered formulations that can form NSVs by hydration prior to adminis- was lower than that of free CPFX. Using the cell viability assay in human
tration [160]. The formation of niosomes using air-jet nebulizers has lung carcinoma cell line (A549), niosome-entrapped CPFX showed
been demonstrated [161,162], however, the actual feasibility, for drug significantly lower cytotoxicity in comparison to the free drug. These
delivery, of niosomes obtained via vibrating-mesh nebulizers has not results indicate that NSVs can be used as carriers for pulmonary delivery
yet been fully investigated. On the other side, proniosomes loaded of CPFX via nebulization [170].
with the anti-asthma steroid BDP were developed to generate niosomes
that are suitable for aerosolization by air-jet or vibrating-mesh 5.5. Parenteral delivery
nebulization.
The Sp 60 niosomes, obtained from the corresponding proniosomes, The parenteral route of administration is one of the most effective
were characterized and their aerosol delivery was investigated using routes for the delivery of the active pharmaceutical substances. To
Omron MicroAir (passively vibrating-mesh) nebulizer, aeroneb pro maintain a therapeutic effective concentration of a drug, it requires
(actively vibrating-mesh) nebulizer and Pari LC print (air-jet) nebulizer. frequent injections which ultimately lead to low patient compliance.
The generated aerosols by all tested devices showed high drug output In parenteral drug delivery, major progresses were obtained in the
and fine particle fraction (FPF). The drug output and FPF of the aeroneb field of vesicular formulation technologies which were in most cases
pro vibrating-mesh nebulizer were similar to those of the standard air- capable to provide a targeted and sustained drug release in predictable
jet nebulizer Pari LC sprint. In vivo investigations still need to explore manner to overcome the problems associated with conventional paren-
the suitability of niosomes as alternatives to liposomes for pulmonary teral delivery systems, especially for drugs with narrow therapeutic
delivery [163]. index and poor bioavailability (Table 7).
Recently Sp and Tw niosomes were studied for the pulmonary deliv- In 2007, Mukherjee et al. presented a study aimed to develop and
ery of anti-tuberculosis drugs [164–168] and of amphotericin B (AMB) compare acyclovir containing nano-vesicular liposomes and niosomes
[169]. AMB is used to treat both fungal and leishmanial infections, based on CHOL, soya L-α-lecithin and the non-ionic surfactant Sp 20.
which are of major significance to human health. Clinical use of free Furthermore, in vitro investigations were performed in order to find
AMB is limited by its nephrotoxicity, whereas liposomal AMB is costly out whether acyclovir-loaded nanovesicles could sustain the release of
and requires parenteral administration. On the other hand, treatment the drug by increasing residence time and thus, acyclovir could reduce
with AMB-loaded niosomes showed a significant reduction in fungal its dose-related systemic toxicity. Obtained results indicated that NSVs
lung burdens in a rat model of invasive pulmonary aspergillosis and in showed better stability compared to liposomes [171].
a significant suppression of Leishmania donovani liver parasite burdens. Nystatin (NYS) is a polyene antifungal drug that has been used in the
The results of this study indicate that AMB aerosolised niosomal formu- treatment of cutaneous, vaginal and oral fungal infections since the
lations enhanced pulmonary and hepatic delivery while minimizing 1950s. NYS encapsulation in niosomes was proposed to obtain a safe
systemic exposure and toxicity [169]. and effective formulation which can be parenterally administered for

Table 7
Niosomes in parenteral delivery.

Compositions Drug Experimental model Results References

Span 60 or Span 40/Chol/SA or DCP. Nystatin (NYS) In vitro release studies, in vivo evaluation of Reduced NYS toxicity and enhanced antifun- [172]
nephrotoxicity and hepatotoxicity, analysis of gal activity, after parenteral administration.
the fungal burden in experimental animals
infected with Candida albicans
Nanohybrid systems of non-ionic Paclitaxel (PTX) In vitro internalization studies (A549 and Empty nanohybrids interfere P-gp expression [178]
surfactant (Solutol® HS 15, pluronic A549/T cells). Effect of blank nanohybrid on the membrane of drug resistant cells and
F68 and cremophor EL) inserting structures on A 549 cells and on P-gp expres- decrease ATP level. Higher cytotoxicity than
liposomes loading paclitaxel sion, on ATP levels. In vitro cytotoxicity ex- liposomes
periment. Apoptosis assay and cell cycle
analysis.
Freeze dried Span 40/Chol niosomes Cisplatin Anticancer efficacy in rats bearing VX2 Inhibition of tumor growth and of tumor [179]
sarcoma metastasis to inguinal lymph nodes and liver,
lower mortality when compared to free drug
Tween 61⁄Chol⁄dodecyl dimethyl Human tyrosinase plasmid Cytotoxicity on B16F10 cells Evaluation of No cytotoxic effect; high stability of the loaded [180]
ammonium (PE) and Tat ⁄human melanin concentration and tyrosinase enzyme plasmid when kept at various temperatures;
tyrosinase plasmid (TPE) activity. enhancement of human tyrosinase gene
expression and melanin production
pH sensitive niosomes 5 Fluorouracil In vitro release studies. In vivo distribution to Remarkable high drug concentration at tumor [181]
tumor sites site
Spans/, Chol/cetyl trimethyl Autoclaved Leishmania major Induction of the immune response against Moderate effect in the prevention of [183,184]
ammonium bromide (ALM) cutaneous leishmaniasis in BALB/c mice cutaneous leishmaniasis in BALB/c mice
200 C. Marianecci et al. / Advances in Colloid and Interface Science 205 (2014) 187–206

neutropenic patients. NYS niosomes were prepared using Sp 60 or Sp and the disappearance of the melting peak of the drug clearly indicated
40 and CHOL. SA and DCP were added as the positive and negative that the drug was encapsulated within NSVs. Tumor-targeted effect
charge-inducing agents, respectively. Neutral and positively charged was proven by the remarkable high concentration of the pH-sensitive
NSVs gave the highest encapsulation efficiencies. The release of neutral niosomes in the tumor site of the mice transplanted with tumor cells.
and negatively charged NYS niosomes was estimated, and it showed The use of NSVs was also evaluated for injectable vaccines formula-
a slow sustained release profile. A 25-kGy γ-irradiation dose was tions. It is known that a promising strategy for overcoming the poor
sufficient to sterilize the investigated vesicles. NYS niosomes exerted immunogenicity of antigen is the development of new vaccine adju-
less nephrotoxicity and hepatotoxicity in vivo, showed higher level vants, or carriers that enhance the effectiveness of vaccines. This occurs
of drug in vital organs and revealed pronounced efficacy in elimination because the adjuvants act as immunostimulatory agents. Given the ever
of the fungal burden in experimental animals infected with Candida increasing interest in vesicle-based vaccines, it is important to under-
albicans compared with those treated with free NYS. Niosomal stand precisely how vesicular carriers interact with the immune system
encapsulation thus provided means for parenteral administration of and stimulate immunity [181]. The first report showing the adjuvant
NYS, reducing its toxicity and making it a more active antifungal agent effect of surfactant vesicles was provided by Brewer and Alexander.
[172]. These authors reported that niosomes are potent adjuvant in terms of
The parenteral route was also proposed for antineoplastic drug immunological selectivity, low toxicity and stability [182]. Sp niosomes
administration. In this case the advantage of drug loading in a surfactant were also proposed as DNA vaccine carriers, after conjugation with OPM
carrier could be the selective delivery of the cytotoxic agent to [183].
the tumor site, thus decreasing toxic effects. Drug loaded vesicles More recently, NSVs were investigated for potential intranasal
could improve the therapeutic outcome and low drug resistance of immunization; the vesicles contained either the secretory recombinant
traditional chemotherapeutics by altering drug pharmacokinetics form of glycoprotein B (gBs) of herpes simplex virus type 1 or a related
and biodistribution. For example intravenous administration of polylysine-rich peptide (DTK) for the induction of protective immunity
hydroxycamptothecin entrapped in niosomes to sarcoma 180 ascites against genital herpes infection in mice [184]. Furthermore, different
tumor bearing mice led to a total regression of the tumor and also higher positively charged niosomal formulations containing Sps, CHOL and
plasma level and slower elimination [173]. cetyl trimethyl ammonium bromide were used for the induction of
Niosomes have great potential in drug nanoformulations for the immune response against cutaneous leishmaniasis in BALB/c mice
melanoma therapy [174–176]. New nanohybrid systems of non-ionic [185].
surfactant and lipid loading paclitaxel (PTX) were prepared to over- Different charged niosomal formulations containing Sps, CHOL and
come multidrug resistance in PTX-resistant human lung cancer cell cetyl trimethyl ammonium bromide were prepared for the entrapment
line. Three non-ionic surfactants, solutol® HS 15, pluronic F68 and of autoclaved Leishmania major (ALM). The selected formulation was
cremophor EL, were inserted into liposomes. The apoptotic assay and used for the induction of the immune response against cutaneous
the cell cycle analysis showed that the nanohybrid systems were able leishmaniasis in BALB/c mice. In vivo studies showed that the niosomes
to induce more apoptotic cells in drug resistant cells when compared containing ALM have a moderate effect in the prevention of cutaneous
with liposomes [177]. Cisplatin-loaded niosomes (CP-NMs) were pre- leishmaniasis in the tested animals [185].
pared using Sp 40 and CHOL as excipients, and then lyophilized and Another interesting goal in niosomal formulations for parenteral
characterized. Their anticancer efficacy was tested in rabbits bearing administration is the possibility to obtain successful targeting strategies,
VX2 sarcoma. The rabbits, locally injected with the CP-NMs, gave signif- avoiding effects on healthy tissues and organs and reaching only
icantly superior results of inhibition of tumor growth, much lower mor- interested area in the body. An emerging paradigm for the design of
tality and improved results of body weight variation and inhibition of effective drug targeting is the modification of vesicular carrier
tumor metastasis to inguinal lymph nodes and liver compared to parameters to encourage the preferential in vivo biodistribution. These
those treated in the same way with the drug solution. The inspiring parameters include size, shape, charge, chemistry, and ligand modifica-
anticancer results indicated that the CP-NMs might be developed as tion. In particular, after intravenous administration, the surfactant
an effective anticancer preparation with low toxicity [178]. vesicles, according to their physic-chemical properties, are surrounded
Potent melanin production enhancement of human tyrosinase plas- by blood components including many types of circulating serum
mid (pAH7⁄Tyr) in mouse melanoma cells (B16F10) by Tat peptide and factors known as opsonins which mark them for clearance by RES
an entrapment in elastic cationic niosomes were described by Manosroi macrophages. For these reasons niosomes can be used for drug targeting
et al. for vitiligo treatment. The NSVs, composed of Tw61, CHOL and in the treatment of diseases in which the infecting organism resides
dodecyl dimethyl ammonium bromide, were prepared by freeze-dried in the RES, exerting a sort of passive targeting. The study of antimony
emptying liposomes method. The B16F10 cells transfected with distribution in mice, performed by Hunter et al. showed high liver
pAH7⁄Tyr –Tat peptide-elastic cationic niosomes (TPE) complexes in level after intravenous administration of the drug carried by NSVs, with-
a specific ratio showed the highest enhancement of the tyrosinase out increasing the damage on heart and kidney [186].
enzyme activity and melanin production. For all TPE complexes a slight More recently, Sp 60/Tw61 niosomes conjugated with a purified
or no cytotoxic effect on the cell line was obtained. The pAH7⁄Tyr- monoclonal antibody to CD44 were proposed for immuno-targeting
loaded elastic cationic niosomes and TPE complexes exhibited high [187] and “hydrid” Tw20 NSVs were reported to show intrinsic selectiv-
stability of the loaded plasmid when kept at various temperatures for ity towards macrophages [91].
8 weeks. This study indicated the enhancement of human tyrosinase Attaching target molecules to niosome surface is one the approach to
gene expression and melanin production with relatively low cytotoxic- overcome the limitation of passive targeting exploiting the EPR effect
ity by Tat and entrapment in elastic cationic niosomes as well as the (e.g. different degree of vascularization and porosity of tumor vessels).
enhancement of stability of the plasmid loaded in the TPE complexes Equipping the surfactant vesicles with a ligand it becomes able to be
[179]. guided to the intended target.
Wang et al. [180] investigated preparation, characterization and Ligands and techniques used to modify niosome surface are similar
tumor-targeted effect of pH-sensitive niosomes, composed of a non- to those used in liposomal formulations [188]: it was reported
ionic surfactant mixed with CHEMS. that glucose and transferrin conjugation of niosomes enhance vesicle
Both normal NSVs and pH-sensitive niosomes showed spherical distribution to the brain or to solid tumors [189,190] and that
morphology under TEM with an appropriate average particle sizes. niosome functionalization by N-palmitoylglucosamine (NPG), allowed
The thermotropic behavior, structure changes and interaction of a reduction of doxorubicin accumulation in the heart, longer blood
5-fluorouracil were characterized by differential scanning calorimetry, circulation times and increased brain concentrations with respect to
C. Marianecci et al. / Advances in Colloid and Interface Science 205 (2014) 187–206 201

the commercial formulation of the drug, after i.v. administration to rats Magneto niosomes, i.e. vesicles based on non-ionic surfactants with
[191]. their aqueous core loaded with both a magnetic material and a model
Although ligand-mediated targeting niosomes have not yet made a drug were proposed by Tavano et al. [200] as theranostic nanocarriers.
considerable progress, it will be feasible and stimulating to expand Magneto NSVs containing doxorubicin were prepared using Tw60 and
researches in this field, taking advantages of knowledge collected in pluronic L64, a copolymer of ethylene oxide and propylene oxide blocks
studying targeting liposomes to overtake their limitations. arranged in a triblock structure, also known, more simply, as pluronic.
Both surfactants present in their structures ethylene oxide chains, that
5.6. Gene delivery are claimed to prevent particle opsonisation, render them ‘unrecogniz-
able’ by RES, and significantly reduce hepatic uptake. Moreover, these
Gene therapies, using genetic material (DNA, oligonucleotides, small moieties were used to sterically stabilize the vesicular carriers and,
interfering RNAs, ribozymes, DNAzymes), rather than traditional drugs hence, to prolong their half-life. The loading of magnetic ferrofluid did
to patients, are being investigated for treatment of a number of different not change the morphology and shape of niosomes (both empty and
inherited or acquired disorders. Unlike traditional pharmaceuticals, drug loaded) and doxorubicin was efficiently encapsulated in both for-
gene therapy has the theoretical potential to treat almost any disease. mulations, due to favorable interactions between magnetic materials
Although gene therapy is believed by several investigators as the and drug. Doxorubicin loading and release behavior of Tw60- and
therapy of the 21st century, numerous barriers still exist that limit the pluronic-based magneto-niosomes could probably promote them as
efficient delivery or expression of genetic material into our body cells effective functional materials for magnetically-controlled cancer therapy.
[192,193]. Different delivery systems are being developed to enhance NSVs prepared using Tw20 or Sp 20 and CHOL loaded with lipophilic
the stability, to increase the efficiency of cellular uptake and intracellu- and hydrophilic magnetic nanoparticles (MNPs) were prepared and
lar trafficking, or to alter the biodistribution of genetic materials. An deeply characterized by Marianecci et al. as promising carriers for
approach involves the use of cationic niosomes as novel non-viral the development of theranostic strategies. In this study the analyzed
carriers for plasmid and oligonucleotide delivery [194,195]. Among formulations confirm the importance of surfactant chemical-physical
various nonionic surfactants, Sps and Tws have been commonly used characteristics in the entrapment of MNPs of different polarity, thus
as additives for the formulation of lipid-based gene carriers. Compared highlighting the high versatility of niosomal bilayers and structures
with Sps, it is worth noting that TWs not only are a critical class of property that make such structures so appealing among drug delivery
emulsifier and stabilizer in those formulations, but they also play an nanocarriers [201].
active role in biological integration with DNA, thereby promoting gene
expression. 6. Natural products delivery
Recently, non-ionic surfactant vesicles, from cationic lipid and Sp 80
were synthesized and evaluated as promising vehicles for small inter- In spite of the high costs related to new drug discovery, there is
fering RNA delivery to efficiently and specifically silence expression of today a great deal of interest in investigating new plant constituents
both green fluorescent protein (66% knockdown) and aromatase (77% and/or new delivery systems for natural products.
knockdown) genes in breast cancer cell lines [196]. As above pointed out, novel delivery systems, such as NSVs, that
Khositsuntiwong et al. investigated the enhancement of tyrosinase modify the pharmacokinetics of phytochemicals can be used to enhance
gene expression and melanin production in tyrosinase gene knocked or modulate the delivery to target sites; consequently the use of NSVs
out human melanoma cells and in B16F10 cells by loading the plasmid was proposed also for several natural products and some of them are
in elastic cationic niosomes [197]. reported (Table 8).
Ginkgo biloba extract (GbE) was investigated for human brain and
5.7. Diagnostics/theranostics breast cancer prevention; GbE entrapped in niosomes (Tw80, Sp 80,
and CHOL) showed an increase of in vivo distribution, after oral admin-
Nanocarrier-mediated delivery has emerged also as a successful istration, when compared to GbE tablet [202].
strategy to enhance delivery of imaging agents for tumors, thereby Niosome formulations loading Gymnema sylvestre extract showed
increasing the potential for diagnosis at an earlier stage. It was showed significant blood glucose level reduction and increased antihyper-
that combination of PEG and glucose conjugates on the surface of glycemic activity compared with the parent extract [203].
niosomes significantly improved tumor targeting of an encapsulated Based on the poor bioavailability of silymarin and on the advantages
paramagnetic agent assessed with nuclear magnetic resonance (NMR) of niosomes, a silymarin niosomal preparation was proposed, showing a
imaging in a human carcinoma xenograft mode [198]. significant decrease in both transaminase levels as well as in serum
Niosomes composed of Tw61, CHOL and DCP were proposed as drug alkaline phosphatase level in comparison with administered silymarin
delivery and imaging in combination with ultrasound. It was evidenced suspension in albino rats [204].
that ultrasound at specific frequencies can reversibly permeabilize the In a recent study the transdermal penetration of papain from gel (8%
lipophilic membrane of niosomes to allow the controlled release of a carbopol®980) formulations containing elastic niosomes (Tw61:CHOL:
compound without destroying the vesicular structure [199]. sodium cholate at 1:1:0.1) and from nanospheres (Poly(lactic-co-

Table 8
Recently proposed pharmaceutical application for natural product-loaded niosomes.

Natural product Niosome formulation Route of administration Reference

Ammonium glycyrrhizinate Sp 20:Tw85:CHOL Tw20:DCP: CHOL Dermal/Transdermal [119,120]


Curcumin Sp 60 or Sp 80 or Tw 20:CHOL Transdermal [208]
Ellagic acid Sp 60 or Tw60:CHOL Transdermal [118]
Ginkgo biloba extract Tw80 or Sp 80:CHOL Oral [202]
Gymnema sylvestre extract Sp 40:CHOL Oral [203]
Papain Tw61:CHOL:sodium cholate Transdermal [205]
Rice bran extract Tw61:CHOL:cetyl trimethylammonium bromide Transfollicular [206,207]
Silymarin Sp 40 or Sp 60:CHOL Subcutaneous [204]
Rutin Sp 60 or Sp 60/Gelucire® Rectal [215]
202 C. Marianecci et al. / Advances in Colloid and Interface Science 205 (2014) 187–206

glycolic acid), PLGA) loaded with papain was compared. The higher po- The increasing number of studies on natural product (Table 8) can
tential of elastic niosomes for the enhancement of rat skin transdermal lead to consider niosomes as promising carriers, by different routes of
absorption of papain and the improvement of scar reduction, when administration [215], in pharmaceutics, in cosmetics as well as for
compared to nanospheres, was demonstrated [205]. food applications.
The physico-chemical properties of cationic niosomes (Tw61/
CHOLl/cetyl trimethylammonium bromide) loaded with rice bran ex- 7. Niosomes vs liposomes
tract were investigated [206]. These carriers appeared to be a suitable
system for topical anti-androgenic alopecia application because of the Taking into account the enormous prospective of vesicular systems
convenient use and high transfollicular penetration in the skin, accom- in drug delivery and targeting, an increasing interest is arising in evalu-
panied by a low systemic effect [207]. ating the possible benefits that niosome entrapment can offer in com-
Different proniosomal gel bases were prepared using Sp 60, Sp 80, parison with liposomes in addition to general considerations as low
Tw20 and CHOL for the encapsulation of curcumin. The investigated costs and higher stability towards oxidative degradation.
formula was not irritant, not toxic, but had lower anti-inflammatory Most of published results concern the ability of vesicles to enhance
and anti-arthritic effects than the marketed indomethacin products percutaneous permeation, thus increasing drug effectiveness after topi-
[208]. cal administration. Formulations with niosomes demonstrated a better
Ellagic acid (EA) is a potent antioxidant phytochemical substance skin permeation potential, sustained release characteristic, and higher
which has limited applications due to its poor biopharmaceutical prop- stability as compared to liposomes.
erties (i.e., low solubility and low permeability). EA loaded niosomes Two mechanism were proposed for the enhancement of permeation
were prepared from Sp 60 and Tw60 mixtures for transdermal delivery. of drugs loaded in niosomes: a permeation enhancer effect and direct
Skin distribution studies evidenced that EA-loaded niosomes were vesicle fusion with the Stratum Corneum (SC) (Fig. 8). This mechanism
more efficient in the delivery of EA through human epidermis and der- is supported by Fang et al. who investigated the feasibility of niosomes
mis than EA solutions [118]. for the transdermal administration of enoxacin and showed that
NSVs made up of Tw85, Sp 20 and CHOL at various concentrations enhancing effect for skin permeation of niosomes was greater than that
were prepared to investigate the possible application of niosomes for of liposomes composed of dimyristoyl phosphatidylcholine (DMPC)
the delivery of ammonium glycyrrhizinate (AG), useful for the treat- [111]. These results were confirmed by several authors for different sur-
ment of various inflammatory-based diseases: the AG-loaded NSVs factant vesicles and different drugs, regardless of the physico-chemical
showed no toxicity, good skin tolerability and were able to improve properties of analyzed nanocarriers: tretinoin [112], resveratrol [216]
the drug anti-inflammatory activity in mice. Furthermore, an improve- and bacitracin zinc [217] in niosomes showed a better behavior than
ment of the anti-inflammatory activity of the niosome delivered drug liposomal formulations in the cutaneous delivery.
was observed on chemically induced skin erythema in humans On the other hand, comparative studies of liposomes and niosomes
[119,120]. for topical delivery of anticancer drug to cancer cells reported opposing
Niosomal formulations were proposed, in preliminary studies, for results. When ciclopirox olamine (CPO) was encapsulated in liposomes
the delivery of colchicine [209], punicalagin [210] and neem seed oil and niosomes, niosomal CPO in comparison to liposomal and pure CPO
[211]. demonstrated cytotoxic effect at significantly lower concentration
Recently, resveratrol was loaded in Sp niosomes to modulate its selectively on cancer cells (KB-oral cancer, PC3-prostate cancer,
release in functional foods [212,213] and a black tea extract/multilamellar Siha-cervical cancer and Vero-kidney epithelial cell lines), improving
niosome formulation (Brij 52:CHOL, 1:1) was investigated as sunscreen the anticancer potential of CPO [218]. On the contrary, in a comparative
agent [214]. study of liposomes and niosomes for topical delivery of 5-fluorouracil

Niosome

(D)
(A) (E)
Drug (C)
(B)

Stratum
corneum

Viable
Epidermis

Dermis

Fig. 8. Possible mechanisms of action of surfactant vesicles for dermal and transdermal applications: (A) drug molecules are released by niosomes, (B) niosome constituents act as
penetration enhancer, (C) niosome adsorption and/or fusion with Stratum Corneum; (D) intact niosome penetration through the intact skin; (E) niosome penetration through hair follicles
and/or pilosebaceous units.
C. Marianecci et al. / Advances in Colloid and Interface Science 205 (2014) 187–206 203

(5-FU) to skin cancer cells (HaCaT), liposomes were found to be more absorption of the drug was reported when compared to oral tablet
cytotoxic on the cell line in comparison with niosomes [219]. administration [231].
Clinical trials by phototrichogram method were performed for
8. Niosome-based patents and pre-clinical and clinical trials proniosomal formulation of finasteride on twenty healthy male volun-
teers and it was found to increase the anagen hair count of test group
In spite of the large number of publications and the wide research by 42.85% when compared with the control group (6.6%), indicating
carried out over the last few decades of niosome formulations and that the proniosomal gel formulation achieved the objective of enhanc-
possible applications, as above reported, only few studies have stepped ing the drug concentration in androgenic receptors of the hair shaft and
into patents and pre-clinical and clinical trials, and these are exclusively thus it can be used safely for treating androgenic alopecia [232].
oriented towards topical delivery. The efficacy of a niosomal gel formulation of griseofulvin in the treat-
About 200 patents concern vesicular carriers, including niosomes, ment of tinea circinata was evaluated on sixteen patients, comparing it
but patents exclusively based on surfactant vesicles are very few and to a griseofulvin gel form and to a placebo gel. The results evidenced an
have been developed in the last five years: most of them reported that effective and safe approach in treating tinea circinata, but underlined
drug inclusion in niosomes has the advantages of improved curative that a further large-scale studies will be needed to establish the higher
effect, reduced adverse side effect and convenient route of administra- efficacy of the niosomal gel formulation [233].
tion. The “niosomal approach” was proposed for different drugs: tacro- Recently, niosomal gel formulations were developed as anti-aging
limus for ophthalmic application after corneal transplantation [220]; agents: niosomes entrapping rice bran bioactive compounds or loaded
beclomethasone for pulmonary delivery [221]; collagen for dermal with a semi-purified fraction from terminalia chebula galls were tested
delivery [222]; metformin for oral delivery [223] and immunogenic on human volunteers by measuring skin elasticity and roughness. In
compounds, including an inactivated virus [224]. both studies a superior clinical anti-aging activity of the niosomal gel
All proposed preparation processes are simple and suitable for com- was reported [234,235].
mercialization and it must be pointed out that the reported applications
for lyophilized niosomal formulation [221,224] could be useful in
further development of surfactant vesicular carriers. 9. Concluding remarks and perspectives
Recent patented niosomal formulation concern surfactant vesicles
with targeting properties exerted by inducing a cholinesterase- Niosomes have been extensively studied as an alternative to
mediated disruption of niosome with time [225] or by surfactant deriv- liposomes. NSVs have some advantages over liposomes, such as their
atization with folic acid [226]. relatively higher chemical stability, improved purity and relatively
Among niosome-based patents also peculiar applications have lower cost in comparison with liposomes. The niosome bilayer is differ-
been proposed such as the preparation of niosome probe for the detec- ent from the liposome bilayer in that NSVs are prepared from uncharged
tion of neomycin with advantages of high selectivity, high sensitivity single-chain surfactants and cholesterol whereas liposomes are
and wide range of application [227] or the possibility to use ultrasound prepared from double-chain phospholipids (neutral or charged) and
to mediate delivery non-invasively by altering the niosome membrane cholesterol. As with liposomes, the in vitro/in vivo properties of
structure [228]. niosomes depend both on the composition of the bilayer and on method
Pre-clinical and clinical trials are exclusively oriented towards of their production. Niosomal drug delivery has yet to make its clinical
dermal and transdermal delivery (Table 9). debut even though these systems have been demonstrated to yield
Lakshmi et al. [129,229] reported the preparation of niosomal meth- promising therapeutics. NSVs alter the plasma clearance kinetics, tissue
otrexate in chitosan gel to be tested for irritation and sensitization on distribution, metabolism and cellular interaction of the drug.
healthy human volunteers and for assessing the efficacy of the gel on Although numerous papers, among those reported in this review,
psoriasis patients. Obtained results suggested that the niosomal metho- show data related to cytotoxicity of niosomal vesicles as well as of
trexate gel is more effective than placebo (as expected) but also of the surfactant monomers; unfortunately, up to date there are no specific
marketed methotrexate gel. wide studies aimed to investigate toxicity of NSVs after in vivo adminis-
NSVs loading urea, dispersed in chitosan gel, were tested on psoria- tration, in particular in long term trials. From published information it is
sis affected patients with less than 25% severity of any category of pso- possible to evaluate the safe concentration of unstructured and struc-
riasis. The niosomal gel produced significant reduction in the lesion than tured surfactants that can be used in cell interaction studies.
the plain urea gel. The niosomal urea gel produced greater reduction in The lack of a specific toxicological study can represent an intriguing
total score and desquamation score compared to erythema and infiltra- opportunity for toxicologists to enrich the knowledge in drug delivery
tion score and proved that niosomal urea in chitosan gel can be used as system tolerability. For example there are no studies on the fate of
an adjuvant in the treatment of psoriasis [230]. surfactant in cells, animals, humans, while the understanding of their
A novel sustained release proniosomal system for transdermal metabolism is a fundamental step in reaching the clinical trials.
delivery of vinpocetine was designed using sugar esters as non-ionic Lipidic vesicles are the first nanomedicine delivery system to make
surfactants in which proniosomes were converted to NSVs upon skin the transition from bench to clinical application, and they are now an
water hydration following topical application under occlusive condi- established technology platform with high clinical acceptance. We can
tions. After in vitro characterization, an optimized formula was clinically look forward to many more efforts in developing surfactant vesicles,
assessed for transdermal pharmacokinetic evaluations and a larger according to the evidence that, despite the many promising proof of

Table 9
Niosomal formulations in pre-clinical and clinical trials (dermal and transdermal delivery).

Active compound Formulation Application Control group Reference

Finasteride Proniosomal gel Androgenetic alopecia Finasteride gel [232]


Griseofulvin Niosomal gel Treatment of tinea circinata Griseofulvin gel and placebo gel [233]
Methotrexate Niosomal gel Psoriasis Marketed methotrexate gel [129,229]
Urea Niosomal gel Psoriasis Urea gel [230]
Vinpocetine Proniosomal gel Cerebrovascular disorders Oral tablet administration [231]
Rice bran bioactive compounds (RBBC) Niosomal gel, niosomal cream Skin aging RBBC gel or cream [234]
Gallic acid Niosomal gel Skin aging Gallic acid gel [235]
204 C. Marianecci et al. / Advances in Colloid and Interface Science 205 (2014) 187–206

concept studies there is still a long road ahead to becoming a clinical [57] Mozafari MR, Method and apparatus for producing carrier complexes. UK Patent
No. GB. 0404993.8, Int. Appl. No. PCT/GB05/000825;2005.
reality. [58] Manosroi A, Chutoprapat R, Abe M, Manosroi J. Int J Pharm 2008;352:248.
In addition, the big chance to the field consists of its highly interdis- [59] Pham TT, Jaafar-Maalej C, Charcosset C, Fessi H. Colloids Surf B Biointerfaces
ciplinary nature from basic chemistry and physic to clinical evaluations. 2012;94:5.
[60] Lo CT, Jahn A, Locascio LE, Vreeland WN. Langmuir 2010;26:8559.
This means there are still many open challenges that need to be [61] Azmin MN, Florence AT, Handjani-Vila RM, Stuart JFB, Vanlerberghe G, Whittaker
overcome and there are still opportunities for academic and industrial JS. J Pharm Pharmacol 1985;37:237.
scientists to make a decisive impact. [62] Wallach DFH. In: Gregoriadis G, editor. Liposome Technology, vol. 2. CRC Press;
1993. p. 141–56.
[63] Vandegriff KD, Wallach DF, Winslow RM. Artif Cells Blood Substit Immobil
References Biotechnol 1994;22:849.
[64] Chopineau J, Lesieur S, Ollivon M. J Am Chem Soc 1994;116:11582.
[1] Peer D, Karp JM, Hong S, Farokhzad OC, Margalit R, Langer R. Nat Nanotechnol [65] Baillie AG, Coombs GH, Dolan TF, Laurie J. J Pharm Pharmacol 1986;38:502.
2007;2:751. [66] Karim KM, Mandal AS, Biswas N, Guha A, Chatterjee S, Behera M. J Adv Pharm
[2] Davis ME, Chen Z, Shin DM. Nat Rev Drug Discov 2008;7:771. Technol Res 2010;1:374.
[3] Kamaly N, Xiao Z, Valencia PM, Radovic-Moreno AF, Farokhzad OC. Chem Soc Rev [67] Duncan R, Florence AT, Uchegbu IF, Cociacinch F, 1997. Drug Polymer conjugates
2012;41:2971. encapsulated within niosomes. International Patent Application PCT: GB97:00072.
[4] Xu ZP, Zeng QH, Lu GQ, Yu B. Chem Eng Sci 2006;61:1027. [68] Manconi M, Sinico C, Fadda AM, Vila AO, Figueruelo J, Molina F. Colloids Surf A:
[5] Bangham AD, Standish MM, Watkins JC. J Mol Biol 1965;13:238. Physicochem Eng Aspect 2005;270–271:124.
[6] Pierre R, Bernadete M, dos Santos Costa MI. Arch Dermatol Res 2011;303:607. [69] Muzzalupo R, Nicoletta FP, Trombino S, Cassano R, Iemma F, Picci N. Colloids Surf B
[7] Al-Jamal W, Kostarelos K. Acc Chem Res 2011;10:1094. Biointerfaces 2007;58:197.
[8] Uchegbu IF, Florence AT. Adv Colloid Interface Sci 1995;58:1. [70] Carafa M, Santucci E, Lucania G. Int J Pharm 2002;231:21.
[9] Uchegbu IF, Vyas SP. Int J Pharm 1998;172:33. [71] Carafa M, Di Marzio L, Marianecci C, Cinque B, Lucania G, Kajiwara K, et al. Eur J
[10] Handjani-Vila RM, Riber A, Rondot B, Valenberghe G. Int J Cosmet Sci 1979;1:303. Pharm Sci 2006;28:385.
[11] Baillie AJ, Florence AT, Hume LR, Muirhead GT, Rogerson A. J Pharm Pharmacol [72] Carafa M, Marianecci C, Lucania G, Marchei E, Santucci E. J Control Release 2004;95:
1985;37:863. 67.
[12] Mozafari MR, Reed CJ, Rostron C. Pharmazie 2007;62:205. [73] Terzano C, Allegra L, Alhaique F, Marianecci C, Carafa M. Eur J Pharm Biopharm
[13] Lesieur S, Madelmont CT, Patenostre M, Moreau JM, Handjani-Vila RM, Ollivon M. 2005;59:57.
Chem Phys Lipids 1990;56:109. [74] Manconi M, Chiara S, Valenti D, Loy G, Fadda AM. Int J Pharm 2002;234:237.
[14] Vanlerberghe G, Morancais JL. STP Pharm Sci 1996;1:5. [75] Manconi M, Valenti D, Sinico C, Lai F, Loy G, Fadda AM. Int J Pharm 2003;260:261.
[15] Jousma H, Joosten JGH, Gooris GS, Junginger HE. Colloid Polym Sci 1989;267:353. [76] Tabbakhian M, Tavakoli N, Jaafari MR, Daneshamouz S. Int J Pharm 2006;323:1.
[16] Jousma H, Joosten JGH, Junginger HE. Colloid Polym Sci 1988;266:640. [77] Jain S, Chaudhari BH, Swarnakar NK. Drug Deliv Transl Res 2011;1:309.
[17] Shahiwala A, Misra A. J Pharm Pharm Sci 2002;5:220. [78] Manvi SR, Gupta VRM, Srikanth K, Devanna N. Res J Pharm Technol 2012;5:1570.
[18] Tavano L, Alfano P, Muzzalupo R, de Cindio B. Colloids Surf B Biointerfaces 2011;87: [79] Vyas SP, Singh RP, Jain S, Mishra V, Mahor S, Singh P, et al. Int J Pharm 2005;296:80.
333. [80] Gupta PN, Mishra V, Rawat A, Dubey P, Mahor S, Jain S, et al. Int J Pharm 2005;293:
[19] Israelachvili JN. In Intermolecular and surface forces. 2nd ed. Academic Press; 1992. 73.
[20] Israelachvili JN, Mitchell DJ, Ninham BW. J Chem Soc Faraday Trans 1976;2:1525. [81] Singh P, Prabakaran D, Jain S, Mishra V, Jaganathan KS, Vyas SP. Int J Pharm
[21] Dai WG, Dong LC, Li S, Pollock-Dove C, Chen J, Mansky P, et al. Int J Pharm 2004;278:379.
2007;336:1. [82] Barenholz Y, Amselem S. Quality control assays in the development and clinical use
[22] Cevc G. J Control Release 2012;160:135. of liposome-based formulations. In: Gregoriadis G, editor. Liposome Technology.
[23] Mohanty A, Dey J. J Chromatogr A 2006;1128:259. 2nd ed. Boca Raton: CRC Press; 1993.
[24] Roy S, Dey J. J Colloid Interface Sci 2007;307:229. [83] Mali N, Darandale S, Vavia P. Drug Deliv Transl Res 2013;3:587.
[25] Morigaki K, Walde P. Curr Opin Colloid Interface Sci 2007;12:75. [84] Liu T, Guo R. Colloid Polym Sci 2007;285:711.
[26] Namani T, Ishikawa T, Morigaki K, Walde P. Colloids Surf B Biointerfaces 2007;54: [85] Liu T, Guo R, Colloids Surfaces A. Colloids Surf A: Physicochem Eng Aspect
118. 2007;295:130.
[27] Wieprecht T, Beyermann M, Seelig J. Biophys Chem 2002;96:191. [86] Cortesi R, Esposito E, Corradini F, Sivieri E, Drechsler M, Rossi A, et al. Int J Pharm
[28] Harvey RD, Heenan RK, Barlow DJ, Lawrence MJ. Chem Phys Lipids 2005;133:27. 2007;339:52.
[29] Dipti S, Tyagi VK. J Oleo Sci 2006;55:215. [87] Binnig G, Rohrer H, Gerber C, Weibel E. Surf Phys Rev Lett 1982;49:57.
[30] Muzzalupo R, Trombino S, Iemma F, Puoci F, La Mesa C, Picci N. Colloids Surf B [88] Zareie MH, Mozafari MR, Hasirci V, Piskin E. J Liposome Res 1997;7:491.
Biointerfaces 2005;46:78. [89] Ramalingam N, Natesan G, Dhandayuthapani B, Perumal P, Balasundaram J,
[31] Muzzalupo R, Ranieri GA, La Mesa C. Langmuir 1996;12:3157. Natesan S. Pak J Pharm Sci 2013;26:1089.
[32] Talsma H, Van Stenberg MJ, Brochert JHC, Crommelin D. J Pharm Sci 1994;83:216. [90] Sennato S, Bordi F, Cametti C, Marianecci C, Carafa M, Cametti M. J Phys Chem B
[33] Bandyopadhyay P, Johnson M. Colloids Surf B Biointerfaces 2007;58:68. 2008;112:3720.
[34] Helenius A, McCaslin DR, Fries E, Tanford C. Methods Enzymol 1979;56:734. [91] Agrati C, Marianecci C, Sennato S, Carafa M, Bordoni V, Cimini E, et al. Nanomed
[35] Carafa M, Santucci E, Alhaique F, Coviello T, Murtas E, Riccieri FM, et al. Int J Pharm Nanotechnol Biol Med 2011;7:153.
1998;160:51. [92] Di Marzio L, Marianecci C, Petrone M, Rinaldi F, Carafa M. Colloids Surf B
[36] Saettone MF, Perini G, Carafa M, Santucci E, Alhaique F. STP Pharm Sci 1996;6:94. Biointerfaces 2011;82:18.
[37] Santucci E, Carafa M, Coviello T, Murtas E, Riccieri FM, Alhaique F, et al. STP Pharm [93] Kreuter J. Colloidal Drug Delivery Systems. Marcel Dekker; 1994 219.
Sci 1996;6:29. [94] Domenici F, Panichelli D, Castellano AC. Colloids Surf B Biointerfaces 2009;69:216.
[38] Nasseri B. Int J Pharm 2005;300:95. [95] Pozzi D, Caminiti R, Marianecci C, Carafa M, Eleonora S, De Sanctis SC, et al. Lang-
[39] Caracciolo G, Pozzi D, Caminiti R, Marianecci C, Moglioni S, Carafa M, et al. Chem muir 2010;26:2268.
Phys Lett 2008;462:307. [96] Lentz BR. Chem Phys Lipids 1993;64:99.
[40] Manconi A, Vila AO, Sinico C, Figueruelo J, Molina F, Fadda AM. J Drug Deliv Sci [97] Sarkar S, Mandal S, Sinha J, Mukhopadhyay S, Das N, Basu MK. J Drug Target
Technol 2006;16:141. 2002;10:573.
[41] Hinz HJ, Kuttenreich H, Mayer R, Renner M, Frund R. Biochemistry 1991;30:5125. [98] Zhai L, Lu X, Chen W, Hu C, Zheng L. Colloids Surf A: Physicochem Eng Aspect
[42] Liu T, Guo R, Hua W, Qui J. Colloids Surf A: Physicochem Eng Aspect 2007;293:255. 2004;236:1.
[43] Manosroi A, Wongtrakul P, Manosroi J, Sakai H, Sugawara F, Yuasa M, et al. Colloids [99] Celia C, Trapasso E, Cosco D, Paolino D, Fresta M. Colloids Surf B Biointerfaces
Surf B Biointerfaces 2003;30:129. 2009;72:155.
[44] Chaw CS, Kim KYA. Pharm Dev Technol 2013;18:667. [100] Manosroi A, Thathang K, Werner RG, Schubert R, Peschka-Süss R, Manosroi J. Int J
[45] Rogerson A, Cummings J, Willmott N, Florence AT. J Pharm Pharmacol 1988;40: Pharm 2008;356:291.
337. [101] Bergstrand N, Arfvidsson MC, Kim J-M, Thompson DH, Edwards K. Biophys Chem
[46] Hu C, Rhodes DG. Int J Pharm 1999;185:23. 2003;104:361.
[47] Junyaprasert VB, Teeranachaideekul V, Supaperm T. AAPS PharmSciTech 2008;9: [102] Masotti A, Vicennati P, Alisi A, Marianecci C, Rinaldi F, Carafa M, et al. Bioorg Med
851. Chem Lett 2010;20:3021.
[48] Heurtault B, Saulnier P, Pech B, Proust JE, Benoit JP. Biomaterials 2003;24:4283. [103] Di Marzio L, Marianecci C, Cinque B, Nazzarri M, Cimini AM, Cristiano L, et al.
[49] Pardakhty A, Shakibaie M, Daneshvar H, Khamesipour A, Mohammadi-Khorsand T, Biochim Biophys Acta 2008;1778:2749.
Forootanfar H. J Microencapsul 2012;29:219. [104] Francis MF, Dhara G, Winnik FM, Leroux JC. Biomacromolecules 2001;2:741.
[50] Oh YK, Kim MY, Shin JY, Kim TW, Yun MO, Yang SJ, et al. J Pharm Pharmacol [105] Chandrababu D, Rayudu NK, Kishorebabu M, Surti N, Patel D. Int J Pharm Sci Rev
2006;58:161. Res 2013;18:176.
[51] Cametti C. Chem Phys Lipids 2008;155:63. [106] Cosco D, Celia C, Cilurzo F, Trapasso E, Paolino D. Expert Opin Drug Deliv 2008;5:
[52] Lasic DD. J Colloid Interface Sci 1990;140:302. 737.
[53] Apoorva A, Neha J, Gauree K, Sachdev Y. Int J Pharm Sci Rev Res 2012;12:53. [107] Trapasso E, Cosco D, Celia C, Fresta M, Paolino D. Expert Opin Drug Deliv 2009;6:
[54] Yasam VR, Jakki SL, Natarajan J, Kuppusamy G. Drug Deliv 2013 [in press]. 465.
[55] Sankar V, Ruckmani K, Durga S, Jailani S. Pak J Pharm Sci 2010;23:103. [108] Paolino D, Muzzalupo R, Ricciardi A, Celia C, Picci N, Fresta M. Biomed Microdevices
[56] Blazek-Welsh AI, Rhodes DG. AAPS PharmSci 2001;3:1. 2007;9:421.
C. Marianecci et al. / Advances in Colloid and Interface Science 205 (2014) 187–206 205

[109] Marianecci C, Rinaldi F, Esposito S, Di Marzio L, Carafa M. Curr Drug Targets [170] Moazeni E, Gilani K, Sotoudegan F, Pardakhty A, Najafabadi AR, Ghalandari R, et al.
2013;14:1070. J Microencapsul 2010;27:618.
[110] Marianecci C, Rinaldi F, Di Marzio L, Ciogli A, Esposito S, Carafa M. Lett Drug Des [171] Mukherjee B, Patra B, Layek B, Mukherjee A. Int J Nanomedicine 2007;2:213.
Discov 2013;10:12. [172] Shafik El-Ridy M, Abdelbary A, Essam T, Mohsen Abd EL-Salam R, Kassem A. Drug
[111] Fang JY, Hong CT, Chiu WT. Int J Pharm 2001;219:61. Dev Ind Pharm 2011;37:1491.
[112] Manconi M, Sinico C, Valenti D, Lai F, Fadda AM. Int J Pharm 2006;311. [173] Shi B, Fang C, Pei Y. J Pharm Sci 2006;95:1873.
[113] Carafa M, Marianecci C, Rinaldi F, Santucci E, Tampucci S, Monti D. J Liposome Res [174] Cosco D, Paolino D, Muzzalupo R, Celia C, Citraro R, Caponio D, et al. Biomed
2009;19:332. Microdevices 2009;11:1115.
[114] Manosroi A, Khanrin P, Lohcharoenkal W, Werner RG, Manosroi J. Int J Pharm [175] Gude RP, Jadhav MG, Rao SG, Jagtap AG. Cancer Biother Radiopharm 2002;17:183.
2010;392:304. [176] Paolino D, Cosco D, Muzzalupo R, Trapasso E, Picci N, Fresta M. Int J Pharm
[115] Sathali AAH, Rajalakshmi G. Int J Pharm Technol Res 2010;2:2081. 2008;353:233.
[116] Gupta NK, Dixit VK. Arch Dermatol Res 2011;303:89. [177] Ji X, Gao Y, Chen L, Zhang Z, Deng Y, Li Y. Int J Pharm 2012;422:390.
[117] El-Laithy HM, Shoukry O, Mahran LG. Eur J Pharm Biopharm 2011;77:43. [178] Yang H, Deng A, Zhang J, Wang J, Lu B. J Microencapsul 2013;30:237.
[118] Junyaprasert VB, Singhsa P, Suksiriworapong J, Chantasart D. Int J Pharm 2012;423: [179] Manosroi J, Khositsuntiwong N, Manosroi W, Götz F, Werner RG, Manosroi A. Drug
303. Deliv 2013;20:10.
[119] Marianecci C, Rinaldi F, Mastriota M, Pieretti S, Trapasso E, Paolino D, et al. J Control [180] Wang M, Yuan Y, Gao Y, Ma HM, Xu HT, Zhang XXN, et al. Drug Dev Ind Pharm
Release 2012;164:17. 2012;38:1134.
[120] Di Marzio L, Marianecci C, Rinaldi F, Esposito S, Carafa M. Lett Drug Des Discov [181] Shilpa S, Srinivasan BP, Chauhan M. Vaccine 2012;30:2256.
2012;9:494. [182] Brewer JM, Alexander J. Immunology 1992;75:570.
[121] Patel KK, Kumar P, Thakkar HP. AAPS PharmSciTech 2012;13:150. [183] Jain S, Vyas SP. J Pharm Pharmacol 2005;57:1177.
[122] Manosroi A, Chankhampan C, Ofoghi H, Manosroi W, Manosroi J. Hum Exp Toxicol [184] Cortesi R, Ravani L, Rinaldi F, Marconi P, Drechsler M, Manservigi M, et al. Int J
2013;32:31. Pharm 2013;440:229.
[123] Ahmed S, Mamdouh G, Shadeed G, Eman M. Int J Drug Dev Res 2013;5:83. [185] Pardakhty A, Shakibaie M, Daneshvar H, Khamesipour A, Mohammadi-Khorsand T,
[124] El-Menshawe SF, Hussein AK. Pharm Dev Technol 2013;18:779. Forootanfar H. J Microencapsul 2012;29:219.
[125] Mishra S, Vasistha P, Sachdeva M, Sara UVS. World J Pharm Pharm Sci 2013;2:554. [186] Brown MD, Schätzlein AG, Uchegbu IF. Int J Pharm 2001;229:1.
[126] Manosroi J, Lohcharoenkal W, Götz F, Werner RG, Manosroi W, Manosroi A. J [187] Marianecci C, Rinaldi F, Di Marzio L, Pozzi D, Caracciolo G, Manno D, et al. Biomed
Pharm Sci 2010;100:1525. Microdevices 2013;15:299.
[127] Antunes FE, Gentile L, Rossi CO, Tavano L, Ranieri GA. Colloids Surf B Biointerfaces [188] Allen TM, Cullis PR. Adv Drug Deliv Rev 2013;65:36.
2011;87:42. [189] Dufes C, Gaillard F, Uchegbu IF, Schatzlein AG, Olievier JC, Muller JM. Int J Pharm
[128] Lakshmi PK, Devi GS, Bhaskaran S, Sacchidanand S. Indian J Dermatol Venereol 2004;285:77.
Leprol 2007;73:157. [190] Hunter CA, Dolan TF, Coombs GH, Baillie AJ. J Pharm Pharmacol 1988;40:161.
[129] Marianecci C, Carafa M, Di Marzio L, Rinaldi F, Di Meo C, Matricardi P, et al. J Pharm [191] Bragagni M, Mennini N, Ghelardini C, Mura P. J Pharm Pharm Sci 2012;15:184.
Pharm Sci 2011;14:336. [192] Hood E, Gonzalez M, Plaas A, Storm J, VanAuker M. Int Pharmacopsychiatry
[130] Kakkar S, Kaur IP. Int J Pharm 2011;413:202. 2007;339:222.
[131] Maheshwari C, Pandey RS, Chaurasiya A, Kumar A, Selvam DT, Prasad GB, et al. Int [193] Hong M, Zhu S, Jiang Y, Tang G, Pei Y. J Control Release 2009;133:96.
Immunopharmacol 2011;11:1516. [194] Huang YZ, Chen JL, Chen XJ, Gao JQ, Liang WQ. J Mater Sci Mater Med 2008;19:607.
[132] Mahor S, Gupta PN, Rawat A, Vyas SP. Curr Med Chem 2007;14:2898. [195] Huang Y, Rao Y, Chen J, Yang VC, Liang W. J Biomed Mater Res A 2011;96:513.
[133] Hamishehkar H, Rahimpour Y, Kouhsoltani M. Expert Opin Drug Deliv 2013;10:261. [196] Zhou C, Mao Y, Sugimoto Y, Zhang Y, Kanthamneni N, Yu B, et al. Mol Pharm
[134] Aggarwal D, Kaur IP. Int J Pharm 2005;290:155. 2012;9:201.
[135] Kaur P, Aggarwal D, Singh H, Kakkar S. Graefes Arch Clin Exp Ophthalmol [197] Khositsuntiwong N, Manosroi A, Götz F, Werner RG, Manosroi W, Manosroi J. J
2010;248:1467. Pharm Pharmacol 2012;64:1376.
[136] Abdelbary G, El-Gendy N. AAPS PharmSciTech 2008;9:740. [198] Luciani A, Olivier JC, Clement O, Siauve N, Brillet PY, Bessoud B, et al. Radiology
[137] Saettone MF, Perini G, Carafa M, Santucci E, Alhaique F. STP Pharm Sci 1996;6:94. 2004;231:135.
[138] Abdelkader H, Ismail S, Kamal A, Alany RG. J Pharm Sci 2011;100:1833. [199] US Patent US20060292211 Ultrasound enhancement of drug release across
[139] Abdelkader H, Wu Z, Al-Kassas R, Alany RG. Int J Pharm 2012;433:142. non-ionic surfactant membranes Hood, Elizabeth; Strom, Joel A.; Vanauker,
[140] Guinedi AS, Mortada ND, Mansour S, Hathout RM. Int J Pharm 2005;306:71. Michael 19 luglio 2011.
[141] Aggarwal D, Pal D, Mitra AK, Kaur IP. Int J Pharm 2007;338:21. [200] Tavano L, Vivacqua M, Carito V, Muzzalupo R, Caroleo MC, Fiore N. Colloids Surf B
[142] Shen Y, Tu J. AAPS J 2007;9:E371. Biointerfaces 2013;102:803.
[143] Benson HAE. Expert Opin Drug Deliv 2006;3:727. [201] Marianecci C, Rinaldi F, Ingallina C, Passeri D, Sorbo A, Rossi M, et al. IL Nuovo
[144] Kaur IP, Rana C, Singh M, Bhushan S, Singh H, Kakkar S. J Ocul Pharmacol Ther Cimento C 2013;36:103.
2012;28:484. [202] Jin Y, Wen J, Garg S, Liu D, Zhou Y, Teng L, et al. Int J Nanomedicine 2013;8:421.
[145] Bayindir ZS, Yuksel N. J Pharm Sci 2010;99:2049. [203] Kamble B, Talreja S, Gupta A, Patil D, Pathak D, Moothedath I, et al. Nanomedicine
[146] Jadon PS, Gajbhiye V, Jadon RS, Gajbhiye KR, Ganesh N. AAPS PharmSciTech 2013;8:1295.
2009;10:1186. [204] El-Ridy MS, Badawi AA, Safar MM, Mohsen AM. Int J Pharm Pharm Sci 2012;4:
[147] Akhter S, Kushwaha S, Warsi MH, Anwar M, Ahmad MZ, Ahmad I, et al. Drug Dev 549.
Ind Pharm 2012;38:84. [205] Manosroi A, Chankhampan C, Manosroi W, Manosroi J. Eur J Pharm Sci 2013;48:
[148] Sankhyan A, Pawar PK. DARU J Pharm Sci 2013;21:7. 474.
[149] Sakthivel M, Kannan K, Manavalan R, Senthamarai R. J Pharm Sci Res 2013;5:8. [206] Manosroi A, Ruksiriwanich W, Masahiko S, Hideki Ak, Manosroi W, Manosroi J. J
[150] Bini DKB, Akhilesh D, Prabhakara P, Kamath JV. Int J Drug Dev Res 2012;4:147. Nanosci Nanotechnol 2012;12:7339.
[151] Di Marzio L, Esposito S, Rinaldi F, Marianecci C, Carafa M. Colloids Surf B [207] Manosroi A, Ruksiriwanich W, Abe M, Manosroi W, Manosroi J. Eur J Pharm
Biointerfaces 2013;104:200. Biopharm 2012;81:303.
[152] Akhilesh D, Faishal G, Prabhu P, Kamath JV. Int J Pharm Pharm Sci 2012;4(Suppl.3): [208] Kumar K, Rai AK. Pharmazie 2012;67:852.
307. [209] Balasubramanian E, Gajendran T. Int J Pharm Bio Sci 2013;4:9.
[153] Nasr M. AAPS PharmSciTech 2010;11:85. [210] Priya H, Singh H. JDDT 2012;2:56.
[154] Xiao Y, Song Y, Chen Z, Ping Q. Int J Pharm 2006;319:162. [211] Chandel A, Saroha K, Nanda S. Int J Pharm Sci Nanotech 2012;5:1780.
[155] Gurrapu A, Jukanti R, Bobbala SR, Kanuganti S, Jeevana JB. Adv Powder Technol [212] Pando D, Gutierrez G, Coca J, Pazos C. J Food Eng 2013;117:227.
2012;23:583. [213] Kamel R, Basha M, Abd El-Alim SH. J Lip Res 2013;23:28.
[156] Yoshida H, Lehr CM, Kok W, Junginger HE, Verhoef JC, Bouwstra JA. J Control [214] Meng-I Yeh MS, Hsiu-Chin H, Jia-Horng L, Min-Chuan H, Kuei-Fen H, Feng-Lin. Int J
Release 1992;1:145. Dermatol 2013;52:239.
[157] Wilkhu JS, McNeil SE, Anderson DE, Perrie Y. J Drug Target 2013;21:291. [215] Agrawal U, Gupta M, Dube D, Vyas SP. Crit Rev Ther Drug Carrier Syst 2013;30:51.
[158] Jain S, Singh P, Mishra V, Vyas SP. Immunol Lett 2005;101:41. [216] Pando D, Caddeo C, Manconi M, Fadda AM, Pazos C. J Pharm Pharmacol 2013;65:
[159] Marianecci C, Paolino D, Celia C, Fresta M, Carafa M, Alhaique F. J Control Release 1158.
2010;147:127. [217] Kasliwal N. J Dispers Sci Technol 2012;33:1267.
[160] Hu C, Rhodes DG. Int J Pharm 2000;206:110. [218] Karimunnisa S, Atmaram P, Shama A, Alpana M. Int J Drug Dev 2012;4:139.
[161] Abd-Elbary A, El-laithy HM, Tadros MI. Int J Pharm 2008;357:189. [219] Alvi IA, Madan J, Kaushik D, Sardana S, Pandey RS, Ali A. Anticancer Drugs 2011;22:
[162] Desai TR, Finlay WH. Int J Pharm 2002;241:311. 774.
[163] Elhissi A, Hidayat K, Phoenix DA, Mwesigwa E, Crean S, Ahmed W, et al. Int J Pharm [220] Xu Y, Li Q. Tacrolimus ophthalmic preparation for corneal transplantation and
2013;444:193. production method thereof by; 2013 [CN 103142468 A 20130612].
[164] Jatav VS, Singh SK, Khatri P, Sharma AK, Singh R. J Chem Pharm Res 2011;3:199. [221] Alhaique F, Carafa M, Fresta M, Marianecci C, Paolino D. Niosomes, freeze-dried
[165] Mehta SK, Jindal N, Kaur G. Colloids Surf B Biointerfaces 2011;87:173. powder thereof and their use in treatment of respiratory disorders by; 2013
[166] Mehta SK, Jindal N. Colloids Surf B Biointerfaces 2013;101:434. [IT 1397274 B1 20130104].
[167] El-Ridy MS, Abdelbary A, Nasr EA, Khalil RM, Mostafa DM, El-Batal AI, et al. Drug [222] Ghulam Nabi Qazi, Farhan Jalees Ahmad, Mohd Samim, Deborah Cooper,
Dev Ind Pharm 2011;37:1110. Marnickavasagar V, Rajendran M. Collagen nanostructures; 2013 [WO 2013015674
[168] Singh G, Dwivedi H, Saraf SK, Saraf SA. Trop J Pharm Res 2011;10:203. A1 20130131].
[169] Alsaadi M, Italia JL, Mullen AB, Ravi Kumar MNV, Candlish AA, Williams RAM, et al. [223] Deng Y, Yang Q, Zhang X. Metformin-containing niosome-like drug delivery system
J Control Release 2012;160:685. and its application by; 2012 [CN 102755292 A 20121031].
206 C. Marianecci et al. / Advances in Colloid and Interface Science 205 (2014) 187–206

[224] Anderson DE. Thermostable lyophilized immunogenic compositions of an [228] Hood E, Strom JA, Vanauker M. Ultrasound enhancement of drug release across
inactivated viral antigen with a non-ionic surfactant vesicle for treating viral infec- nonionic surfactant membranes; 2013 [US 8435558 B1 20130507].
tions; 2013 [WO 2013104995 A2 20130718]. [229] Lakshmi PK, Bhaskaran S. Int J Pharm Sci Rev Res 2011;7:1.
[225] Liu Y, Wang K, Guo D, Wang Y. Cholinesterase-regulated nanosize supermolecular [230] Mazda F, Özer AY, Ercan MT. STP Pharm Sci 1997;7:205.
niosome, its preparation method and application; 2012 [CN 102580100 A 20120718]. [231] Vimal KR, Sankar V, Srinivas CR, Kumaresan M. Indian Drugs 2012;49:47.
[226] Yang H, Huang J, Deng A, Wu X, Wan Z, Hu Q, et al. Folic acid-modified cisplatin- [232] Kassem MAA, Esmat S, Bendas ER, El-Komy MHM. Mycoses 2006;49:232.
targeting non- ionic niosome and its preparation method; 2012 [CN 102552132 [233] de Souza AL, Kiill CP, dos Santos FK, da Luz GM, Rocha e Silva H, Chorilli M, et al.
A 20120711]. Curr Nanosci 2012;8:512.
[227] Chen X, Zhou G, Wang H. The invention disclosed a process for prepn. of niosome [234] Manosroi A, Jantrawut P, Akihisa T, Manosroi W, Manosroi J. Pharm Biol 2011;49:1190.
probe used for detecting Neomycin; 2013 [CN 103232358 A 20130807]. [235] Manosroi A, Chutoprapat R, Abe M, Manosroi W, Manosroi J. Pharm Biol 2012;50:208.

Das könnte Ihnen auch gefallen