Advanced Drug Delivery Reviews 55 (2003) 519–548
www.elsevier.com / locate / addr
Poly(alkylcyanoacrylates) as biodegradable materials for
biomedical applications
Christine Vauthier*, Catherine Dubernet, Elias Fattal, Huguette Pinto-Alphandary,
Patrick Couvreur
´
Laboratoire de Physico-chimie, Pharmacotechnie et Biopharmacie, UMR CNRS 8612, Universite de Paris XI,
´
5 Rue Jean Baptiste Clement, 92296 Chatenay-Malabry Cedex, France
Received 26 September 2002; accepted 30 January 2003
Abstract
This review considers the use of poly(alkylcyanoacrylates) (PACAs) as biomedical materials. We first present the different
aspects of the polymerization of alkylcyanoacrylate monomers and briefly discuss their applications as skin adhesives,
surgical glues and embolitic materials. An extensive review of the developments and applications of PACAs as nanoparticles
for the delivery of drugs is then given. The methods of preparation of the nanoparticles are presented and considerations
concerning the degradation, in vivo distribution, toxicity and cytotoxicity of the nanoparticles are discussed. The different
therapeutic applications are presented according to the route of administration of the nanoparticles and include the most
recent developments in the field.
2003 Elsevier Science B.V. All rights reserved.
Keywords: Poly(alkylcyanoacrylates); Polymerization; Nanoparticles; Degradation; Therapeutic applications
Contents
1. Introduction ............................................................................................................................................................................
2. Polymerization of alkylcyanoacrylates and biomedical applications of the monomers ...................................................................
2.1. Polymerization of alkylcyanoacrylates ...............................................................................................................................
2.2. Biomedical applications....................................................................................................................................................
3. Poly(alkylcyanoacrylate) nanoparticles......................................................................................................................................
3.1. Preparation of poly(alkylcyanoacrylate) nanoparticles .........................................................................................................
3.1.1. Preparation of nanospheres by emulsion polymerization............................................................................................
3.1.2. Preparation of nanocapsules by interfacial polymerization .........................................................................................
3.1.3. Synthesis of block copolymers and preparation of nanoparticles by nanoprecipitation and emulsification–solvent
evaporation ............................................................................................................................................................
3.2. Degradation of poly(alkylcyanoacrylate) nanoparticles ........................................................................................................
3.3. In vivo distribution of nanoparticles after intravenous administration ...................................................................................
3.4. Toxicity and cytotoxicity of poly(alkylcyanoacrylate) nanoparticles .....................................................................................
*Corresponding author. Fax: 1 33-1-4661-9334.
E-mail address: christine.vauthier@cep.u-psud.fr (C. Vauthier).
0169-409X / 03 / $ – see front matter 2003 Elsevier Science B.V. All rights reserved.
doi:10.1016 / S0169-409X(03)00041-3
520
520
520
520
522
522
522
524
525
526
526
528
520
C. Vauthier et al. / Advanced Drug Delivery Reviews 55 (2003) 519–548
3.5. Therapeutic applications of poly(alkylcyanoacrylate) nanoparticles ......................................................................................
3.5.1. Intravenous administration ......................................................................................................................................
3.5.1.1. Application to the treatment of intracellular infections..................................................................................
3.5.1.2. Application to the treatment of non-resistant cancers ....................................................................................
3.5.1.3. Application to the treatment of resistant cancers ..........................................................................................
3.5.1.4. Application to the delivery of oligonucleotides ............................................................................................
3.5.1.5. Application to the passage of the blood–brain barrier ...................................................................................
3.5.2. Oral route ..............................................................................................................................................................
3.5.2.1. Oral delivery of peptides, proteins and vaccines ...........................................................................................
3.5.2.2. Bioadhesive nanoparticles ..........................................................................................................................
3.5.2.3. Application to the administration of antiproteases ........................................................................................
3.5.3. Other routes of administration .................................................................................................................................
4. Conclusion..............................................................................................................................................................................
References ..................................................................................................................................................................................
1. Introduction
Poly(alkylcyanoacrylates) (PACAs) were not employed as polymers until the early 1980s [1]. However,
the
corresponding
monomers,
alkylcyanoacrylates, have been used since at least
1966 because of their excellent adhesive properties
resulting from the bonds of high strength they are
able to form with most polar substrates, including
living tissues and skin [2]. Therefore, the monomers
have been used extensively as tissue adhesives for
the closure of skin wounds [3–7], as surgical glue,
and as embolitic material for endovascular surgery
[6,8]. More recently, one application of the polymers
consists of the use of PACAs as drug nanoparticulate
carriers [9–15]. This very exciting area of research,
which emerged in the 1980s [1,16,17], has gained
increasing interest in therapeutics, especially for
cancer treatments, which generally involve highly
toxic molecules in contact with healthy tissue. Other
molecules of interest, including poorly stable compounds such as peptides and nucleic acids, have been
combined with PACA nanoparticles for targeting
purposes [12,13,18]. Today, PACA nanoparticles are
considered the most promising polymer colloidal
drug delivery system and are already in clinical
development for cancer therapy [19–23].
This review thus considers, in detail, PACAs as
materials for biomedical applications. The different
aspects presented include a short summary of the
polymerization and biomedical applications of the
monomers and an extensive review of the more
recent developments and applications of PACAs
applied as nanoparticles for drug delivery.
531
531
531
533
535
536
537
538
539
540
541
541
541
542
2. Polymerization of alkylcyanoacrylates and
biomedical applications of the monomers
2.1. Polymerization of alkylcyanoacrylates
The monomers generally occur as clear and colorless liquids with a low viscosity and are highly
reactive compounds, extremely difficult to handle in
their pure form [2,4,23–25]. They display a remarkable tendency to polymerize because of their strong
reactivity. Inhibitors are essential to maintain their
stability. Indeed, alkylcyanoacrylates are able to
polymerize extremely rapidly in the presence of
moisture or traces of basic components.
The polymerization of alkylcyanoacrylates can
theoretically occur according to three different mechanisms, namely free-radical, anionic and zwitterionic
polymerization. In practice, the anionic and zwitterionic routes are strongly favored because they are
rapidly initiated at ambient temperature (Fig. 1).
Classical initiators of anionic polymerization are
anions (i.e. I 2 , CH 3 COO 2 , Br 2 , OH 2 , etc.), weak
bases such as alcohols and water and amino acids
encountered in living tissues [2]. Initiation of polymerization by the amino acids of proteins is responsible for the strong binding power of these components to the skin, as will be explained below.
2.2. Biomedical applications
Alkylcyanoacrylates have been used for decades
as adhesives, mainly for consumer applications
where speed of cure is needed. These monomers
have also been applied in the biomedical area as
C. Vauthier et al. / Advanced Drug Delivery Reviews 55 (2003) 519–548
521
Fig. 1. Chemical structure of alkylcyanoacrylates (A) and scheme of their anionic and zwitterionic polymerization (B).
tissue
adhesives
since
1964,
when
methylcyanoacrylate was first tested for the closure
of 3-cm-long cystotomies in dogs. At present, use as
a tissue adhesive is an improved and popular method
of wound closure, being faster, less painful and more
economical than suturing. It is mainly applied to the
closure of simple lacerations and surgical wounds
[5,7,26,27]. Today, methylcyanoacrylate (Biobond)
is still in use in Japan [28], whereas early
cyanoacrylates were removed from distribution in
other countries because of rapid in vivo degradation,
resulting in significant tissue toxicity and inflammation [27]. They were replaced by longer-chain
alkylcyanoacrylates such as N-butylcyanoacrylate
(Indermil , liquiband ), which is used clinically in
Europe, Canada and the USA, and octylcyanoacrylate (Dermabond ), which received
Food and Drug Administration approval in 1998 and
is now marketed in the USA for skin wound closure
after lacerations or incisions [27]. Upon application,
the liquid monomer formulation polymerizes instantaneously into a thin polymer film that adheres
tenaciously to the mucosal tissue. The polymer film
also creates a mechanical barrier which maintains a
natural healing environment for the area to heal [29].
Octylcyanoacrylate forms strong bonds across opposed wound edges and provides a flexible waterresistant coating, inhibiting microbial growth, there-
522
C. Vauthier et al. / Advanced Drug Delivery Reviews 55 (2003) 519–548
fore avoiding the occurrence of infections [30]. This
new adhesive presents advantages over nbutylcyanoacrylate, including a higher breaking
strength, flexibility and resistance to splintering after
drying [30]. Octylcyanoacrylate also appears to be a
stronger tissue adhesive than its corresponding fibrin
sealant counterpart [7]. Some authors claim that
complications are virtually non-existent [26]. No
evidence of histotoxicity has been reported with this
monomer, which is considered a promising alternative to the standard wound closure method, providing a faster repair of traumatic lacerations and
surgical incisions with similar cosmetic outcomes
[29]. Furthermore, this method of wound closure is
readily accepted by patients, especially by children.
One disadvantage of the current octylcyanoacrylate
formulation is its slow rate of biodegradation. It has
also been stressed that these adhesives are only for
external use.
The internal use of octylcyanoacrylate has not yet
been approved and applications beyond the skin are
considered by certain authors as unwise and potentially dangerous to patients [7]. Despite this,
alkylcyanoacrylates are under investigation for many
other applications [28]. They are even the main
liquid adhesives under clinical investigation for use
in the vascular system as embolitic material and are
being considered to play an important role in managing abnormalities, especially in arteriovenous malformations [6,8]. For instance, Trufill n-BCA , a
combination of n-butylcyanoacrylate and tantalum
powder, is delivered through the arterial system to
stop bleeding atrioventricular malformations and is
then removed during repair. Another active area of
research is the treatment of gastric varices, for which
cyanoacrylate glues are still first choice [31].
3. Poly(alkylcyanoacrylate) nanoparticles
Nanoparticles is a general word used to designate
small-sized polymer particles with a diameter ranging from several tens to several hundred nanometers.
It includes colloidal particles of different structures:
nanospheres, nanocapsules, core-shell nanospheres
and core-shell nanocapsules. The PACA nanoparticle
family comprises nanospheres, oil- and water-containing nanocapsules and core-shell nanospheres
Fig. 2. Schematic representation of the different types of PACA
nanoparticles produced.
(Figs. 2 and 3). PACA nanoparticles can be prepared
either by polymerization of alkylcyanoacrylate
monomers or directly from the polymers (see Section
3.1, Fig. 4). These nanoparticles were developed in
order to design biodegradable drug carrier systems
for targeting a drug to tissues, cells or subcellular
compartments, as explained in Sections 3.2–3.5.
3.1. Preparation of poly(alkylcyanoacrylate)
nanoparticles
3.1.1. Preparation of nanospheres by emulsion
polymerization
Emulsion polymerization is a very popular approach used to synthesize polymer colloids with a
matrix structure (nanospheres, Fig. 2a). The emulsion
polymerization of alkylcyanoacrylates was first introduced by Couvreur et al. in 1979 [1] to design
nanoparticles with biodegradable polymers for the in
vivo delivery of drugs. The polymerization media
used for the polymerization of alkylcyanoacrylates
for this purpose are usually very complex. In these
systems, the polymerization is initiated by the hydroxyl ions of water, and elongation of the polymer
chains occurs according to an anionic polymerization
mechanism (Fig. 1). It should be pointed out that the
anionic polymerization of such a reactive monomer
can be controlled in an aqueous medium. The main
parameters involved are the adjustment of the pH
with a strong mineral acid such as hydrochloric acid
and the concentration of the anionic polymerization
inhibitor (SO 2 ) in the monomer [32]. The size of the
nanospheres produced can vary from 50 to 300 nm
[33–35]. As an example of this procedure, the
monomer (100 ml) is dispersed in acidified water
containing a surfactant or a stabilizing agent (10 ml
C. Vauthier et al. / Advanced Drug Delivery Reviews 55 (2003) 519–548
523
Fig. 3. Example electron micrographs of PACA nanoparticles obtained under different conditions. (A) Oil-containing nanocapsules after
staining with uranyl acetate. (B) Oil-containing nanocapsules after staining with phosphotungstate acid. (C) Water-containing nanocapsules
after cryofracture and shadowing with platinum. (D) Nanospheres after staining with uranyl acetate. (E) Nanospheres after cryofracture and
shadowing with platinum (bars 200 nm; the arrow in cryofractures (C) and (D) indicates shadowing orientation).
of a 0.5–1% solution of Pluronic F68 or dextran 70
at pH 2.5 adjusted with HCl) and allowed to
polymerize spontaneously for a few hours (3–4 h)
under strong magnetic stirring.
Many drugs can be entrapped in PACA nanospheres [14,33]. The addition of cyclodextrins to the
polymerization medium can promote the association
of poorly water-soluble drugs with the PACA nanospheres [36].
It should be pointed out that certain drugs are
reported to be able to initiate the polymerization of
alkylcyanoacrylates, leading to loss of their biological activity [37–39]. However, such side re-
actions could also be used advantageously to promote the covalent binding of certain compounds to
the nanospheres when stable association is required.
Naphthalocyanines [40], a series of photosensitizers
used in the phototherapy of tumors, and a series of
molecules containing diethyltriaminepentacetic acid
(DTPA) capable of complexing radioactive metals
for radiolabelling of nanoparticles in medical imaging [41], have been associated with nanospheres
using this approach. These reactions have also been
used to produce nanospheres with modified surface
properties, allowing the covalent coupling of macromolecules onto the nanosphere surface [33,42–45].
524
C. Vauthier et al. / Advanced Drug Delivery Reviews 55 (2003) 519–548
Fig. 4. Summary of the different methods developed for the preparation of PACA nanoparticles with the corresponding types of
nanoparticles produced.
3.1.2. Preparation of nanocapsules by interfacial
polymerization
Methods based on interfacial polymerization have
been developed to prepare nanocapsules consisting
of a liquid core surrounded by a thin polymer
envelope [46–50]. The reactions are performed
either in water-in-oil or in oil-in-water emulsion
systems or in microemulsions, leading to the production of water-containing or oil-containing
nanocapsules, respectively.
Oil-containing nanocapsules are obtained by the
polymerization of alkylcyanoacrylates at the oil /
water interface of a very fine oil-in-water emulsion
[46]. An organic phase (oil, 1 ml; isobutyl-2cyanoacrylate, 0.125 ml; drug dissolved in ethanol or
acetone, 25 ml) is injected into the aqueous phase
(50 ml) containing a hydrophilic surfactant
(Pluronic F68, 0.25%) under strong magnetic stirring. The nanocapsules form immediately to give a
milky suspension. The organic solvent is then evaporated. In such a system, the organic solvent, acetone
or ethanol, which is totally miscible with water,
serves as a monomer vehicle and the interfacial
polymerization of alkylcyanoacrylate is believed to
occur at the surface of the oil droplets that form
during emulsification [12,51–53]. To promote
nanocapsule formation, an ideal oil / ethanol ratio of
2% in the organic phase has been suggested [51] and
the use of aprotic solvents such as acetone and
acetonitrile has been recommended [54]. Protic
solvents, such as ethanol, n-butanol and iso-propanol, were found to induce the formation of nanospheres in addition to nanocapsules.
Nanocapsules prepared by this method contain
more than 90% oil by weight [53], allowing the
efficient encapsulation of soluble oily substances
[55]. Because of the extremely fast formation of the
polymer shell around the oil droplets, highly watersoluble molecules such as insulin can also be encapsulated with high encapsulation yields (up to
97%) if these compounds are suspended in the oily
phase [52,56–58].
Water-containing nanocapsules may be obtained
by
the
interfacial
polymerization
of
alkylcyanoacrylate in water-in-oil microemulsions.
In these systems, water-swollen micelles of surfac-
C. Vauthier et al. / Advanced Drug Delivery Reviews 55 (2003) 519–548
525
tants of small and uniform size are dispersed in an
organic phase. The monomer is added to the microemulsion and polymerizes at the surface of the
micelles. The polymer forms locally at the water–oil
interface and precipitates to produce the nanocapsule
shell [47,49,50]. Nanocapsules obtained by this
method are of special interest for the encapsulation
of water-soluble molecules such as peptides [50] and
nucleic acids, including antisense oligonucleotides
[49]. For intravenous administration, aqueous corecontaining nanocapsules can be transferred into an
aqueous continuous phase by ultracentrifugation of
the oily suspension over a layer of pure water
containing Span 80 [49].
3.1.3. Synthesis of block copolymers and
preparation of nanoparticles by nanoprecipitation
and emulsification–solvent evaporation
A major problem with colloidal drug carriers after
their intravenous administration is their non-specific
uptake by the macrophages of the mononuclear
phagocyte system. In an attempt to reduce particle
interactions with blood opsonins, which facilitate
phagocytosis, the development of sterically stabilized
nanoparticles with amphiphilic block copolymers has
been introduced, leading to a new generation of drug
carrier systems. These particles remain in the blood
circulation for a longer period of time and their
accumulation in the mononuclear phagocyte system
organs is reduced. To obtain such long-circulating
(Stealth ) nanoparticles, (PACA)–poly(ethylene glycol) (PEG) copolymers have been synthesized using
two methods. With the first method, block copolymers are obtained by initiating the polymerization of
alkylcyanoacrylates either by monomethoxy (MeO)PEG-triphenylphosphine or by triphenylphosphinePEG-triphenylphosphine, leading to diblock MeOPEG–PACA or triblock MeO-PACA–PEG–PACAOMe linear copolymers, respectively (Fig. 5A and
B) [59]. The second route of synthesis is based on
the Knoevenagel reaction of block copolymers,
involving the condensation of a cyanoacetate derivative with formaldehyde [60]. This route leads to
branched copolymers in which the PEG part constitutes the branches of the copolymer (Fig. 5C). This
second method is preferred since it allows the
production of very stable nanospheres and avoids the
use of the triphenylphosphine group, which may be
Fig. 5. Structure of amphiphilic PACA-containing block copolymers: PEG-triphenylphosphate–PACA diblock (A) and triblock
(B) copolymers [59], and monomethoxy or monoamino poly(PEGCA-co-HDCA) copolymer (C) [20,60].
toxic [61–63]. In addition, Stella et al. [20] reported
the preparation of PEG-coated nanospheres with an
amino-terminated PEG-containing copolymer, which
were found to be excellent supports for coupling
ligands at the nanoparticle surface to develop
nanoparticles for cell-selective targeting of drugs.
This approach has been used with folic acid for the
targeting of KB cancer cells.
Nanospheres are prepared from these copolymers
by nanoprecipitation and emulsification–solvent
evaporation. Compared with the methods based on
polymerization, these approaches have the major
advantage that the polymers entering the composition
of the nanoparticles are well characterized and their
intrinsic physico-chemical characteristics do not
depend on the conditions used during the preparation
of the nanospheres. Because of the amphiphilic
nature of these block copolymers, they self-organize
to produce core-shell nanospheres with a hydrophilic
surface (Fig. 2).
Nanoprecipitation is a method based on the formation of colloidal polymer particles during phase
separation induced by the addition of a non-solvent
of the polymer to a rather dilute polymer solution. At
that stage, the particles form spontaneously and
quasi-instantaneously. The polymer solvent is then
526
C. Vauthier et al. / Advanced Drug Delivery Reviews 55 (2003) 519–548
removed from the suspension by rotoevaporation. To
facilitate the formation of colloidal polymer particles
during the first step of the procedure, phase separation is performed with a totally miscible solvent
which is also a non-solvent of the polymer.
The second method, called emulsification–solvent
evaporation, also includes two steps. The first consists of the emulsification of the polymer solution in
an aqueous phase with the aid of a high-pressure
homogenizer or microfluidizer to produce emulsion
droplets of very small size. During the second step,
the polymer solvent is evaporated, inducing polymer
precipitation as nanospheres.
These methods for the preparation of nanoparticles
have been applied to poly(poly(ethylene glycol)
cyanoacrylate-co-hexadecylcyanoacrylate)
(poly(PEGCA-co-PHDCA)) to prepare PEG-coated
nanospheres [20–23,60,64,65].
3.2. Degradation of poly(alkylcyanoacrylate)
nanoparticles
The degradation and toxicity of PACA are often
discussed in the literature, especially for in vivo
applications of PACA nanoparticles as drug delivery
systems. Indeed, the suitability of a polymer designed for use as a drug carrier system for humans
requires that the material has to be biocompatible,
possibly biodegradable, or at least should be able to
be excreted (e.g., by the kidneys). These aspects are
discussed below.
PACA are bioerodible polymers for which complete excretion of the polymer material will only
occur if the nanoparticles were designed using lowmolecular-weight polymers. Indeed, even if different
pathways for PACA degradation have been described
in the literature, the predominant mechanism greatly
depends on the surrounding conditions. One of the
degradation mechanisms described in the literature
consists of the hydrolysis of the ester bond of the
alkyl side chain of the polymer (Fig. 6A) [66–68].
Degradation products consist of an alkylalcohol and
poly(cyanoacrylic acid), which are soluble in water
and be eliminated in vivo via kidney filtration. This
degradation has been shown to be catalyzed by
esterases from serum, lysosomes and pancreatic juice
[69,70] and is believed to occur as the major
degradation pathway in vivo. According to this
mechanism, nanoparticles are usually degraded within a couple of hours depending on the alkyl side
chain length of the PACA forming the nanospheres
[2,67,71].
Another mechanism which may also theoretically
occur in biological systems consists of an unzipping
depolymerization of the parent polymer with immediate repolymerization to give a new polymer with a
much smaller molecular weight. The whole phenomenon occurs within a few seconds and is generally
induced by a base (Fig. 6B) [25]. However, this
mechanism, which may theoretically be induced in
vivo by the amino acids of proteins, has never been
described. Due to its very rapid occurrence, it is
expected that it will be extremely difficult to observe, especially in complex systems such as biological media.
The well-known inverse Knoevenagel reaction,
resulting in the production of formaldehyde and
cyanoacetic ester, has also been reported, but is
limited in water and at physiological pH, yielding
only 5% degradation after 24 h. This mechanism has
been claimed to occur in vivo in the early stage of
the development of PACA for biomedical applications; it was even believed to be responsible for the
toxic effects of the nanoparticles [2,72–75]. However, this degradation pathway is too slow to compete with the other, much more rapid, mechanisms
occurring in vivo catalyzed by enzymes [73,76,77].
3.3. In vivo distribution of nanoparticles after
intravenous administration
The main attraction of PACA nanoparticles is their
ability to achieve tissue targeting and enhance the
intracellular penetration of drugs. After intravenous
administration, PACA nanoparticles are taken up by
the liver, by the spleen and, to a smaller extent, by
bone marrow [78]. Within tissues, nanoparticles are
mainly taken up by macrophages of the mononuclear
phagocyte system [79,80]. However, the architecture
of the organs has been shown to play a role in the
localization of the nanospheres. For instance, in the
spleen of mice, uptake was mainly observed in
metallophilic macrophages of the marginal zone,
whereas, in the rat, which has a sinusoidal spleen
similar to that of humans, particles were found in the
red pulp macrophages [81]. The uptake of nanoparti-
C. Vauthier et al. / Advanced Drug Delivery Reviews 55 (2003) 519–548
527
Fig. 6. Degradation pathways of poly(alkylcyanoacrylate). (A) Enzymatic degradation catalyzed by esterases and leading to the production
of alkylalcohol and poly(cyanoacrylic acids) [66–69]. (B) Unzipping depolymerization–repolymerization mechanism producing oligomers
of PACA [25].
cles by macrophages occurs via an endocytosis
process (Fig. 7), after which the particles end up in
the lysosomal compartment [79] where they are
degraded, producing soluble, low-molecular-weight
compounds that are then eliminated from the body
by renal excretion [66]. Due to their strong lysosomal localization, one can imagine that nanoparticles
are not suitable for the targeting of drugs to the
cytoplasm of cells. To avoid their entrapment within
the lysosomal compartment, several compounds able
to destabilize the lysosomal membrane are added to
the nanoparticulate system (e.g. cationic surfactants)
[82], allowing some drugs to be delivered to the cell
cytoplasm.
528
C. Vauthier et al. / Advanced Drug Delivery Reviews 55 (2003) 519–548
Fig. 7. Endocytosis of PACA nanospheres by J774 macrophages in culture cells as observed by transmission electron microscopy. (A)
Interactions of nanospheres with the cell membrane (arrows). (B) Endocytosis of a nanosphere. (C) Nanospheres in phagosomes (arrows).
Coating PACA nanospheres with PEG results in a
lower uptake of nanoparticles by the mononuclear
phagocyte system and a longer circulation time in the
blood [63,83,84]. As a consequence, these so-called
‘stealth ’ nanoparticles would be able to extravasate
across the endothelium, which becomes permeable
due to the presence of solid tumors. However, from
the point of view of targeting, these ‘stealth ’
nanoparticles are simple and passive systems with no
specific targeting ligands. They basically exploit both
the differences in microvascular permeability between healthy and altered tissues and their longcirculating properties.
3.4. Toxicity and cytotoxicity of
poly(alkylcyanoacrylate) nanoparticles
Nanoparticles are generally captured by the
macrophages of the mononuclear phagocyte system
after intravenous administration. However, depending on their surface characteristics, nanoparticles
may be more or less opsonized, so that coating
nanoparticles with hydrophilic polymers results in a
significant modification of the body distribution
profile. It is evident that the particle distribution
profile in the body may influence the toxicity of the
polymer–drug entity. Indeed, the altered phar-
C. Vauthier et al. / Advanced Drug Delivery Reviews 55 (2003) 519–548
macokinetics and disposition of drugs when associated with nanoparticles should induce novel modalities of cells, tissues or receptor exposures, as well
as novel drug metabolism and drug interactions. For
instance, in some cases, alteration of the drug
distribution profile by linkage to nanospheres could
considerably reduce the toxicity of a drug because of
the decrease in drug accumulation in organs where
the most acute toxic effects are exerted. This concept
was illustrated with doxorubicin, which displays
severe acute and chronic cardiomyopathy. After
intravenous administration to mice, plasma levels of
doxorubicin were higher when the drug was adsorbed onto nanospheres and, at the same time, the
cardiac concentration of the drug was dramatically
reduced [85]. In accordance with the observed
distribution profile, doxorubicin associated with
nanospheres was found to be less toxic than free
doxorubicin [86].
On the contrary, if the cardiac toxicity of doxorubicin is clearly reduced after linkage with PACA
nanoparticles [86,87], increased bone marrow toxicity may be observed [80]. Thus, the evaluation of
potential novel toxicities of polymeric site-specific
drug delivery systems includes the search for a
depression or activation of the mononuclear
phagocyte system. As far as the mononuclear
phagocyte system is concerned, it was observed that
PACA nanoparticles did not induce a physical blockade of this tissue after repeated administration,
although a temporary depletion of blood opsonins
was observed [88]. For this route of administration,
hemocompatibility also needs to be evaluated in
terms of embolies due to particle aggregation or
hemolysis due to erythrocyte damage by nanoparticles or their degradation products. For other means
of administration (transmucosal, oral, regional administration, etc.), it is especially important to determine the absence of local tissue irritation (histocompatibility). In this case, the test of lesion
reversibility is very important to evaluate the acceptability of the nanoparticle system. On the other
hand, as reported by Ammoury et al. [89], the
encapsulation of drugs such as indomethacin into
PACA nanocapsules may also protect mucosae
against the ulcerative properties of the drug.
Results from the toxicological evaluation of PACA
529
nanoparticles motivated the beginning of clinical
trials for human cancer [19,90]. A phase I trial
confirmed the good tolerance of the drug carrier,
since only secondary effects due to the associated
drug were observed.
Cytotoxicity is another important parameter to
consider with nanoparticulate drug delivery systems
since they can be phagocytosed by cells. As reported
by Maassen et al. [91], the cytotoxicity of nanospheres may be due to many factors, such as (1) the
release of degradation products, (2) the stimulation
of cells and subsequent release of inflammatory
mediators, and (3) membrane adhesion. The first
factor, cytotoxicity attributed to the presence of
degradation products, has been reported by several
authors. Kante et al. [92] incubated PACA nanospheres with macrophages, and morphological cell
changes were characterized by electronic microscopy. Only in the presence of poly(methylcyanoacrylate) was cell membrane perforation observed. By contrast, no morphological
changes
were
observed
with
poly(butylcyanoacrylate). Another study [93] was carried
out by incubating nanoparticles with hepatocytes.
Cell toxicity was assessed using a dye-exclusion test
and by measuring lactate dehydrogenase leakage.
Poly(butylcyanoacrylate) nanospheres did not induce
any cytotoxicity at a concentration of 75 mg / ml.
Nevertheless, membrane damage appeared at a polymer concentration of 150 mg / ml. These data were
confirmed by incubating poly(isobutylcyanoacrylate)
nanospheres with L929 fibroblasts, where no cell
mortality was induced after 3 and 7 h incubation at a
polymer concentration of 100 mg / ml. However, after
24 and 48 h, an increased mortality rate was observed [71]. Kubiak et al. [94] have shown that
poly(isobutylcyanoacrylate) nanospheres are more
cytotoxic than poly(isohexylcyanoacrylate) by
measuring [ 3 H]leucine incorporation in resistant and
sensitive cancer cells (DC3F). Gipps et al. [95]
studied the interaction of PACA nanoparticles with
mesenchymal malignant and normal cells. Using
nanospheres made with PACA of different side-chain
length
(poly(methylcyanoacrylate),
poly(ethylcyanoacrylate), poly(butylcyanoacrylate), poly(isobutylcyanoacrylate),
poly(isohexylcyanoacrylate)), the authors showed that the cell viability, as
530
C. Vauthier et al. / Advanced Drug Delivery Reviews 55 (2003) 519–548
determined by the uptake of neutral red, was influenced dramatically by the nature of the polymer.
Polymers with short alkyl side chains appeared more
toxic than polymers with longer alkyl groups, whereas cytotoxicity was found to be independent of the
cell type used. Morphological changes in the cell
membrane were also characterized by transmission
electron
microscopy.
With
poly(isobutylcyanoacrylate), a loss of adhesion followed
by dilation of the rough endoplasmic reticulum of the
cells was observed. Perforation of the cell membrane
occurred later in the damage sequence. The toxicity
of PACA nanospheres was also investigated by
measuring cell growth inhibition of Swiss 3T3
fibroblasts. Poly(isobutylcyanoacrylate) and poly(ethylcyanoacrylate) were found to inhibit cell proliferation to a lower extent than poly(ethoxy-ethyl-2cyanoacrylate) and poly(methylcyanoacrylate) nanospheres. The extent of cell growth inhibition decreased with increasing molecular weight of the
polymer [75]. In addition, Lherm et al. [71] have
shown that, after incubating PACA nanoparticles
with L929 fibroblasts, only nanoparticles with slow
degradation kinetics (i.e. long alkyl side chain) were
non-toxic. From these studies, it can be concluded
that the cytotoxicity of alkylcyanoacrylate polymers
is clearly dependent upon the length of the alkyl side
chain, with a very low toxicity for the longer alkyl
side chains. This observation may be related to the
burst release of degradation products during the
incubation of poly(methylcyanoacrylate) or poly(ethylcyanoacrylate) nanoparticles, since the hydrolytic degradation of poly(alkylcyanoacrylates) increases with decreasing alkyl chain length [2,69].
A second mechanism due to membrane adhesion
was also found to be involved in the cytotoxicity of
PACA nanoparticles. Even though polymers with
longer alkyl chains are less toxic, poly(ethylcyanoacrylate) nanoparticles are more cytotoxic
to L929 fibroblasts than poly(methylcyanoacrylate)
nanoparticles. The reason for this is that poly(methylcyanoacrylate) is very rapidly degraded in the
culture medium and does not adhere to the cell
membrane. On the contrary, poly(ethylcyanoacrylate)
is more cytotoxic because the particles probably first
adhere to the cell membrane and then release their
biodegradation products locally [71].
Finally, the release of inflammatory mediators
induced by the contact of PACA nanoparticles with
cells was studied by Gaspar et al. [96], showing that
PACA nanoparticles could stimulate macrophages.
The release of hydrogen peroxide from the cells was
observed, whereas no release of cytokines could be
detected.
It should be stressed that one has to be very
careful with toxicity studies performed in vitro
because the nanoparticles-to-cell ratio involved under
such conditions is dramatically higher than it would
be after in vivo administration. In addition, if
degradation products are partially or totally responsible for the cytotoxicity of PACA polymers, under
in vivo conditions these products are generally
eliminated from their site of degradation, thus the
contact time with the cells would be considerably
lower than in vitro.
In vitro cell culture experiments are clearly useful
to clarify the polymer–cell interaction, but they are
of poor toxicological relevance without being complemented by ex vivo and in vivo assays. This was
the reason why ex vivo models were used by
Fernandez-Urussuno et al. [97,98] to study the
possible alteration of hepatocytes after repeated
administration of PACA nanoparticles to rats. After
treatment, rat hepatocytes were isolated and both
secretion of inflammation proteins (a-1 acid
glycoprotein) and the oxidative response were observed. However, these effects were reversible after
treatment with PACA nanospheres was discontinued.
Such an hepatocyte reaction was assumed to be due
¨
to the release of mediators from Kupffer cells, in
which the nanoparticles concentrated after intravenous administration. This hypothesis was confirmed
by showing that particles coated with a hydrophilic
¨
block copolymer (therefore escaping Kupffer cell
uptake) do not induce any inflammatory response
[97].
Finally, from a toxicological point of view, it is
noteworthy that the association of a drug with
nanoparticles can also induce a higher intracellular
concentration of the drug and even allows nonintracellularly diffusible drugs to concentrate in
certain intracellular compartments (lysosomes) [99].
This approach, which has been used to improve the
efficacy of certain drugs, may induce unexpected
toxicity in the case of compounds with an apparent
low cytotoxicity because they usually diffuse poorly
C. Vauthier et al. / Advanced Drug Delivery Reviews 55 (2003) 519–548
intracellularly. This was demonstrated with propidium iodide, free or associated with polymeric
nanoparticles [100]. In addition, the pathway and
velocity of cellular absorption can influence the
intracellular distribution of the drug–polymer association, with, as a consequence, altered cytotoxicity.
3.5. Therapeutic applications of
poly(alkylcyanoacrylate) nanoparticles
Polymer nanoparticles are very promising drug
delivery systems for a wide range of applications.
Among the different biodegradable nanoparticles
developed over the last 25 years, an important part of
the published literature has been devoted to PACA
nanoparticles, which have been tested for different
therapeutic purposes and routes of administration.
3.5.1. Intravenous administration
3.5.1.1. Application to the treatment of intracellular
infections
Intracellular infections have been established as a
field of interest for drug delivery by means of
nanospheres. Indeed, infected cells may constitute a
‘reservoir’ for microorganisms which are protected
from antibiotics inside lysosomes. The resistance of
intracellular infections to chemotherapy is often
related to the low uptake of commonly used antibiotics or to their reduced activity at the acidic pH of
lysosomes. To overcome these effects, the use of
ampicillin, a b-lactam antibiotic, bound to PACA
nanospheres was proposed as an endocytozable
formulation [101]. The effectiveness of poly(isohexylcyanoacrylate) nanospheres was tested in the treatment of two experimental intracellular infections.
First, ampicillin-loaded nanospheres were tested in
the treatment of experimental Listeria monocytogenes infection in congenitally athymic nude mice, a
model involving chronic infection of both liver and
spleen macrophages [102]. After adsorption of ampicillin onto nanospheres, the therapeutic activity of
the ampicillin was found to increase dramatically
over that of the free drug. Bacterial counts in the
liver were reduced at least 20-fold after linkage of
ampicillin to poly(isohexylcyanoacrylate) nanospheres. In addition, ampicillin-loaded nanoparticles
531
were capable of ensuring liver sterilization after two
injections of 0.8 mg of nanosphere-bound drug,
whereas no such sterilization was observed with any
of the other regimens tested. The reappearance of
living bacteria in the liver after cessation of treatment was probably due to a secondary infection
derived from other organs such as the spleen, which
was not completely sterilized by the treatment [102].
Secondly, nanosphere-bound ampicillin was tested
in the treatment of experimental salmonellosis in
C57 / BL6 mice, a model involving an acute fatal
infection [101]. All mice treated with a single
injection of nanoparticle-bound ampicillin (dose 0.8
mg) survived, whereas all control mice and all those
treated with unloaded nanospheres died within 10
days post-infection. With free ampicillin, an effective
cure required three doses of 32 mg each. Lower
doses (3 3 0.8 mg and 3 3 16 mg) delayed, but did
not reduce, mortality. Thus, the therapeutic index of
ampicillin, calculated on the basis of mice mortality,
was increased by 120-fold when the drug was bound
to nanospheres.
In order to clarify the mechanism by which PACA
nanospheres improve the antimicrobial efficacy of
ampicillin, Forestier et al. [103] compared the efficacy of ampicillin bound to poly(isobutylcyanoacrylate) nanospheres with that of free ampicillin in
terms of survival of L. monocytogenes in mouse
peritoneal macrophages cultivated in vitro. After 30
h incubation, nanosphere-bound ampicillin reduced
the number of viable bacteria by 99% as compared to
the controls, whereas with free ampicillin the number
of bacteria was slightly lower than in the controls.
Nanoparticle–ampicillin thus appears to be much
more effective than free ampicillin for inhibiting the
intracellular growth of L. monocytogenes.
With in vitro Salmonella typhimurium-infected
macrophages, the situation was slightly more complicated since the bactericidal effect of ampicillinbound poly(isohexylcyanoacrylate) nanospheres was
poor, although the intracellular capture of ampicillin
was dramatically increased and its efflux in the
extracellular medium reduced [104]. In another
study, confocal microscopy and transmission electron microscopy were used to establish the intracellular trafficking of ampicillin-bound poly(isohexylcyanoacrylate) nanospheres and its relation to
bacteria within subcellular compartments (Fig. 8)
532
C. Vauthier et al. / Advanced Drug Delivery Reviews 55 (2003) 519–548
Fig. 8. Transmission electron microscopy of J774 macrophages infected with Salmonella typhymurium incubated with ampicillin-loaded
PACA nanospheres. (A) Colocalization of nanospheres (arrows) and a dividing bacterium (b) in the same vaccuole inside the macrophage.
(B) Ultrastructural autoradiograph of a macrophage treated with [ 3 H]ampicillin-loaded PACA nanospheres. Silver grains (arrows) due to the
presence of radioactive material ([ 3 H]ampicillin) were located inside the cells on spherical bodies that may correspond to bacteria in the
form of spheroplasts, because of the action of the antibiotic, a b-lactam, on the outer membrane, resulting in a loss of rigidity.
[105]. Data obtained from this study clearly demonstrated the active uptake by phagocytosis of ampicillin-bound poly(isohexylcyanoacrylate) nanospheres
by murine macrophages and their localization in the
same vacuoles as the infecting bacteria, but in a
restrictive
way
[105].
Furthermore,
using
[ 3 H]ampicillin-loaded nanospheres, the radioactivity
due to ampicillin could clearly be localized as being
associated with the membrane of intracellular bacteria under lysis due to the presence of the antibiotic
(Fig. 8B). It is difficult to understand the limited
bactericidal effect of ampicillin-bound nanospheres
measured in vitro in this model. The most probable
explanation may be found in one of the known
antibiotic resistance mechanisms of S. typhimurium
involving the inhibition of phagosome–lysosome
fusion required for the nanospheres (in phagosomes)
to reach the bacteria (in lysosomes) [106]. If this
hypothesis is correct, the high efficiency observed in
vivo would be due to the specific targeting of the
infected tissues (rich in macrophages), rather than to
efficient intracellular targeting, as was assumed.
In order to eliminate both dividing and non-dividing bacteria, a fluoroquinolone antibiotic, ciprofloxacin,
has
been
associated
with
poly(isobutylcyanoacrylate) and poly(isohexylcyanoacrylate) nanospheres. In an animal model of persisting Salmonella infection, although an effect on the
early phase of the infection was observed, neither
free nor nanosphere-bound ciprofloxacin was able to
eradicate truly persisting bacteria [107].
Since they accumulate in the mononuclear
phagocyte system, nanospheres also hold promise as
drug carriers for the treatment of visceral leis-
C. Vauthier et al. / Advanced Drug Delivery Reviews 55 (2003) 519–548
hmaniosis [108]. Thus, poly(isohexylcyanoacrylate)
nanospheres were used as a carrier of primaquine,
the activity of which was increased 21-fold against
intracellular Leishmania donovani when associated
with nanospheres [96]. A part of the activity was
attributed to the fact that the phagocytosis of nanospheres lead to the induction of a respiratory burst
which was more pronounced in infected than in
non-infected macrophages [96]. Dehydroemetine is
another drug candidate for this treatment. The heart
toxicity of this drug can be reduced after linkage
with nanospheres [109].
3.5.1.2. Application to the treatment of non-resistant
cancers
When given intravenously, anticancer drugs are
distributed throughout the body as a function of the
physico-chemical properties of the molecule. A
pharmacologically active concentration is reached in
the tumor tissue at the expense of massive contamination of the rest of the body. For cytostatic compounds, this poor specificity raises a toxicological
problem which represents a serious obstacle to
effective therapy. The use of colloidal drug carriers
could represent a more rational approach to specific
cancer therapy. In addition, the possibility of overcoming multi-drug resistance might be achieved by
using cytostatic-loaded nanospheres.
The anti-tumor efficacy of doxorubicin-loaded
PACA nanospheres was first tested using the
lymphoid leukemia L1210 as a tumor model. In this
study, one intravenous injection of doxorubicinloaded poly(isobutylcyanoacrylate) nanospheres was
found to be more effective against L1210 leukemia
than the drug administered in its free form following
the same dosing schedule [87]. However, although
the increased life-span of mice injected with doxorubicin-loaded poly(isobutylcyanoacrylate) nanospheres was twice as long as the increased life-span
for free doxorubicin, there were no long-term survivors. The effectiveness of doxorubicin-loaded
poly(isohexylcyanoacrylate) nanospheres against
L1210 leukemia was even more pronounced than
that
of
doxorubicin
loaded
onto
poly(isobutylcyanoacrylate) nanospheres. The drug toxicity was markedly reduced, so that impressive results
were obtained at doses for which the therapeutic
efficiency of free doxorubicin was completely
533
masked by the overpowering toxicity of the drug
[87]. Preliminary experiments suggested that one
intravenous bolus injection of doxorubicin-loaded
nanospheres was more active, in L1210-bearing
mice, than perfusion of the free drug for 24 h.
The superiority of doxorubicin targeted with the
aid of PACA nanospheres was later confirmed in a
murine hepatic metastases model (M5076 reticulosarcoma) [110]. Irrespective of the dose and the
administration schedule, the reduction in the number
of metastases was much greater with doxorubicinloaded nanospheres than with free doxorubicin,
particularly if treatment was given when the metastases were well established. The improved efficacy
of the targeted drug was clearly confirmed by
histological examinations showing that both the
number and the size of the tumor nodules were
smaller when doxorubicin was administered in its
nanoparticulate form [110]. Furthermore, liver biopsies of animals treated with the nanosphere-targeted
drug showed a lower cancer cell density inside tumor
tissue. Necrosis was often less widespread with the
nanosphere-associated drug than in the control group
and the group treated with free doxorubicin. Studies
performed on total homogenates of livers from both
healthy and metastases-bearing mice showed extensive capture of nanoparticulate doxorubicin by the
liver; no difference in hepatic concentrations was
observed between healthy and tumor-bearing animals
[110]. In order to elucidate the mechanism behind
the enhanced efficiency of doxorubicin-loaded
nanospheres, doxorubicin measurements were made
in both metastatic nodules and neighboring healthy
hepatic tissue. This provided quantitative information
concerning the drug distribution within these tissues
[86]. During the first 6 h after administration,
exposure of the liver to doxorubicin was 18 times
greater for nanosphere-associated doxorubicin. However, no special affinity for the tumor tissue was
detected and the nanospheres were observed by
¨
electron microscopy to be located within Kupffer
cells (macrophages). However, at later time points,
the amount of drug in the tumor tissue increased in
nanosphere-treated animals to 2.5 times the level
found in animals given free doxorubicin. Since
uptake of nanospheres by neoplastic tissue appeared
to be unlikely, this increase in the doxorubicin
concentration in tumor tissue probably resulted from
534
C. Vauthier et al. / Advanced Drug Delivery Reviews 55 (2003) 519–548
Fig. 9. Mechanism proposed to explain the action of doxorubicin-loaded PACA nanospheres in the treatment of liver metastasis. (A) Uptake
¨
of the nanospheres by Kupffer cells. (B) Degradation of the doxorubicin-loaded nanospheres in the intralysosomal compartments of the
¨
Kupffer cells, inducing the release of the drug and diffusion into the surrounding tissue.
doxorubicin released from healthy tissue, in par¨
ticular Kupffer cells (Fig. 9). Hepatic tissue could
play the role of a drug reservoir, from which
prolonged diffusion of the free drug (from nanos¨
pheres entrapped in Kupffer cell lysosomes) towards
neighboring malignant cells occurs (Fig. 9).
This hypothesis raises the question of the longterm effect of an 18-fold increase in the doxorubicin
concentration in the liver. Although toxicological
data have shown that doxorubicin-loaded nanospheres are not significantly or unexpectedly toxic to
the liver in terms of survival rate at high doses, body
weight loss and histological appearance [111], this
possibility should be borne in mind, especially since
¨
a temporary depletion in the number of Kupffer
cells, and hence the ability to clear bacteria, was
observed in rats treated with doxorubicin-loaded
liposomes [112]. Nanoparticle-associated doxorubicin also accumulates in bone marrow, leading to a
myelosuppressive effect [80]. This tropism of carriers might be useful for the delivery of myelostimulating compounds such as granulocyte colony
stimulating factor to reverse the suppressive effects
of intense chemotherapy [113].
Another approach to cancer therapy considered
PEG-coated PACA nanoparticles as a long-circulating carrier for the targeting of recombinant tumor
necrosis factor-a (rHuTNF-a) to tumor tissue
[23,64]. rHuTNF-a was successfully associated with
PEG-coated nanospheres consisting of poly(PEGCAco-PHDCA) [23,64]. The pharmacokinetics and antitumor effect were evaluated in vivo in mice with
sarcoma-180 cells implanted intradermally. As expected because of the difference in microvascular
permeability between healthy and tumoral tissue,
greater accumulation of rHuTNF-a in the tumor and
increased antitumoral activity were found when this
compound was injected intravenously as PEG-coated
nanospheres compared to the free drug [23].
To develop cell-selective targeting, folic acid has
been conjugated to PEG-coated nanospheres consisting of poly(PEGCA-co-PHDCA) [20], the rational
behind this construction being that folic acid-binding
protein is frequently overexpressed on the surface of
human cancer cells. The nanospheres were prepared
using a copolymer containing PEG with a terminal
amino group. This amino group was exposed at the
particle surface and subsequent coupling with activated folic acid was successful. Based on plasmon
resonance assays, it was found that the folate-conjugated nanospheres interacted much more with folatebinding protein than nanospheres coated with PEG.
Folate-decorated nanospheres additionally showed a
greater affinity for the receptor compared to a single
molecule of folic acid. This can be explained by the
cooperative interactions obtained with the nanos-
C. Vauthier et al. / Advanced Drug Delivery Reviews 55 (2003) 519–548
pheres [20]. Folic acid-decorated nanospheres offer
interesting perspectives for the selective targeting of
anticancer compounds to tumoral cells and tissues.
3.5.1.3. Application to the treatment of resistant
cancers
The ability of tumor cells to develop simultaneous
resistance to multiple lipophilic compounds is a
major problem in cancer chemotherapy. Cellular
resistance to anthracyclines has been attributed to an
active drug efflux from resistant cells linked to the
presence of transmembrane P-glycoprotein, which is
not detectable in the parental drug-sensitive cell line.
Drugs such as doxorubicin appear to enter the cell by
passive diffusion through the lipid bilayer. Upon
entering the cell, these drugs bind to P-glycoprotein,
which forms transmembrane channels and uses
energy from ATP hydrolysis to pump these compounds out of the cell [114]. To solve this problem,
many authors have proposed the use of competitive
P-glycoprotein inhibitors, such as the calcium channel blocker verapamil, which are able to bind to
P-glycoprotein and overcome pleiotropic resistance.
However, since verapamil exhibits serious adverse
effects, its clinical use to overcome multidrug resistance is limited. This is why the effect of nanospheres
loaded with doxorubicin, the resistance to which is
known to be related to the presence of P-glycoprotein, was evaluated. The cytotoxicity of free doxorubicin,
doxorubicin-loaded
poly(isohexylcyanoacrylate) nanospheres (mean diameter 300 nm) and
nanospheres without drug against sensitive (MCF7)
and multi-drug resistant (doxorubicin R MCF7)
human breast cancer cell lines was compared [115].
MCF7 cells were more sensitive to free doxorubicin
than doxorubicin R MCF7 cells with a 150-fold
difference in the IC 50 values. No significant difference was observed in the survival rate of MCF7 cells
treated with free doxorubicin or doxorubicin-loaded
nanospheres. In contrast, for doxorubicin R MCF7,
the IC 50 for doxorubicin was 130-fold lower when
doxorubicin-loaded nanospheres were used instead of
free doxorubicin [115]. These results indicate that
PACA nanospheres provide an effective carrier for
introducing a cytotoxic dose of doxorubicin into the
pleiotropic resistant human cancer cell line Dox R
MCF7.
535
Complementary experiments, conducted with
other sensitive and resistant cell lines, have confirmed this efficacy of nanospheres [116,117]. Doxorubicin resistance was circumvented in the majority
of cell lines tested, and encouraging results were
obtained in vivo in a P388 model growing as ascites
[116]. Further studies were undertaken to elucidate
the mechanism of action of PACA nanospheres. The
incubation time and number of particles per cell were
important factors [118] and, when poly(isobutylcyanoacrylate) nanospheres were used,
doxorubicin accumulation within P388 /ADR-resistant leukemic cells increased compared with the free
drug, although no endocytosis of nanospheres
occurred [119]. On the other hand, when the less
rapidly degradable poly(isohexylcyanoacrylate)
nanospheres were used, reversion was observed in
the absence of increased intracellular drug concentration [120]. The degradation products of PACA
nanospheres (mainly poly(cyanoacrylic acid)) were
also able to increase both the accumulation and
cytotoxicity of doxorubicin, although they were
soluble in the culture medium. Hence, reversion of
resistance seems to be due both to the adsorption of
nanospheres on the cell surface and to the formation
of a doxorubicin–poly(cyanoacrylic acid) complex
(an ion pair) which facilitates the transport of the
drug across the cell membrane (Fig. 10) [120,121].
In the light of the results obtained with doxorubicin-loaded PACA nanospheres in the liver metastases
model described above [110], the role of macrophages as a reservoir for doxorubicin was tested in a
two-compartment, co-culture system in vitro with
both resistant and sensitive P388 cells [122]. Even
after prior uptake by macrophages, doxorubicinloaded poly(isobutylcyanoacrylate) nanospheres were
able to overcome resistance. However, this reversion
was only partial. It was decided to take advantage of
the particulate drug carrier effect to associate an
anti-cancer drug and a compound capable of inhibiting P-glycoprotein. This approach was tested with
doxorubicin and cyclosporin A bound to the same
nanospheres, and was found to be extremely effective in reversing P388 resistance [123]. The association of cyclosporin A with nanospheres would ensure
that it reaches the same sites as the anti-cancer drug
at the same time and would also reduce its toxic
side-effects.
536
C. Vauthier et al. / Advanced Drug Delivery Reviews 55 (2003) 519–548
Fig. 10. Proposed mechanism for the reversion of multidrug resistance by means of doxorubicin-loaded PACA nanospheres. Given as free
drug, doxorubicin is pumped out of the cell by P-glycoprotein (A). When given as PACA nanospheres, doxorubicin and degradation products
such as poly(cyanoacrylic acid) are released locally from the nanospheres adsorbed on the cell membrane. Together they can form ion pairs,
which facilitate the penetration of doxorubicin into resistant cells (B) [120].
3.5.1.4. Application to the delivery of oligonucleotides
Oligodeoxynucleotides are potentially powerful
new drugs because of their selectivity for particular
gene products in both sense and anti-sense strategies.
However, using oligonucleotides in therapeutics is a
challenge to pharmaceutical technology because of
their susceptibility to enzymatic degradation and
their poor penetration across biological membranes.
Thus, nanoparticle preparations might be an interesting alternative for protecting oligonucleotides from
degradation in biological fluids. In the case of PACA
nanospheres, since oligonucleotides have no affinity
for the polymeric matrix, association has been
achieved by ion pairing using a cationic surfactant,
cetyltrimethylammonium bromide (CTAB) or diethylaminoethyl-dextran (DEAE), adsorbed onto the
nanosphere surface [82,124].
The oligonucleotides bound to the nanospheres
were protected from nuclease degradation in vitro
[82] and their intracellular uptake was increased
[125]. In addition, PACA nanospheres were able to
concentrate intact oligonucleotides in the liver and
spleen [126]. Antisense oligonucleotides formulated
in this way are also able to specifically inhibit
mutated Ha-ras-mediated cell proliferation and
tumorigenicity in nude mice [127].
However, these nanospheres have two drawbacks:
(i) their toxicity, mainly due to the presence of
CTAB [128]; and (ii) the rapid desorption of the
oligonucleotides in the presence of serum, which
results from PACA nanosphere surface erosion by
serum esterases [126]. This is why PACA nanocapsules with an aqueous core containing the oligonucleotides were developed recently [49,128].
Stability studies demonstrated that the PACA
nanocapsules were able to protect the oligonucleotides from degradation by serum nucleases and that
this protection was more efficient than that obtained
with CTAB-coated PACA nanospheres [82,129].
Phosphorothioate oligonucleotides directed against
EWS Fli-1 chimeric RNA were encapsulated within
PACA nanocapsules and tested for their efficacy in
vivo against experimental Ewing sarcoma in mice
after intratumoral administration [130]. Only intratumoral injection of antisense-loaded nanocapsules
lead to a significant inhibition of tumor growth at a
cumulative dose of 14.4 nmol. No antisense effect
could be detected with the free oligonucleotide. In a
previous study, using the same antisense sequence as
C. Vauthier et al. / Advanced Drug Delivery Reviews 55 (2003) 519–548
the free drug, Tanaka et al. [131] demonstrated
inhibition of tumor growth in a similar model, but a
cumulative dose of 500 nmol oligonucleotide was
needed. With PACA nanocapsules it was possible to
obtain a comparable effect with a 35-fold lower
dose. Therefore, nanocapsule technology allows
smaller phosphorothioate doses to be used and thus
avoid the toxicity and loss of specificity resulting
from phosphorothioates at higher doses [132].
The mechanism by which oligonucleotides in
nanocapsules lead to a significant effect on tumor
growth may be explained by the protection of the
oligonucleotide afforded by the nanocapsules which,
in addition, may act as a controlled release system
for the oligonucleotide within the tumor. Thus, the
use of phosphorothioates at low doses combined with
nanocapsules may represent a new and safe option
for the administration of antisense oligonucleotides
in vivo.
3.5.1.5. Application to the passage of the blood–
brain barrier
The blood–brain barrier is an insuperable obstacle
for a large number of drugs, such as antibiotics,
antineoplastic agents, and a variety of central nervous system-active drugs, especially neuropeptides.
PACA nanoparticles coated with polysorbate 80 have
been proposed to overcome this barrier and to deliver
drugs to the brain [133]. Drugs that have been
transported successfully into the brain using this
carrier include the hexapeptide dalargin [134], the
dipeptide kytorphin [133], loperamide [135],
tubocurarine [136], the NMDA receptor antagonist
MRZ 2 / 576 [137,138], and doxorubicin [139,140].
PACA nanoparticles may be especially helpful for
the treatment of disseminated and very aggressive
brain tumors. Intravenously injected doxorubicinloaded, polysorbate 80-coated nanoparticles lead to a
40% cure in rats with intracranially transplanted
glioblastomas 101 / 8 [139,140]. The mechanism of
nanoparticle-mediated transport of drugs across the
blood–brain barrier has not been fully elucidated.
The most likely mechanism is endocytosis followed
by transcytosis by the endothelial cells lining the
blood capillaries of the brain, as shown by fluorescence labelling and confocal laser scanning microscopy observations [141,142]. Nanoparticle-mediated
drug transport to the brain depends on coating the
537
particles with polysorbates, especially polysorbate 80
[143]. Polysorbate 80-coated PACA nanospheres
were found to adsorb apolipoprotein E from blood
plasma and therefore may mimic low density
lipoprotein, having specific receptors at the surface
of endothelial cells of the blood–brain barrier. After
such recognition, the drug may be released into these
cells from the nanospheres and diffuse into the brain
interior, or the particles may be transcytosed. Other
processes such as tight junction modulation or Pglycoprotein inhibition may also occur (Fig. 11).
Moreover, these mechanisms may run in parallel or
may be cooperative, thus enabling efficient drug
delivery to the brain. However, Olivier et al. [144]
have shown that non-specific permeabilization of the
blood–brain barrier, probably related to the toxicity
of the carrier, may account for the central nervous
system drug penetration when associated with poly(butylcyanoacrylate) nanoparticles and polysorbate
80.
Calvo et al. [22,145] evaluated the ability of longcirculating, PEG-coated PACA nanoparticles consisting of the amphiphilic copolymer poly(PEGCA-coHDCA) to diffuse into brain tissue after intravenous
administration to mice and rats. Based on their longcirculating characteristics, PEG-coated nanoparticles
penetrated into the brain to a greater extent than all
the other nanoparticle formulations tested, including
polysorbate 80-coated nanospheres. Particles were
localized in the ependymal cells of the choroid
plexus, in the epithelial cells of the pia mater and
ventricles, and to a smaller extent in the capillary
endothelial cells of the blood–brain barrier. These
phenomena occurred without any modification of
blood–brain barrier permeability, whereas polysorbate 80-coated nanoparticles owe their efficacy, in
part, to blood–brain barrier permeabilization induced
by the surfactant. Poloxamine 908-coated nanoparticles, which also exhibit long-circulating properties,
failed to increase the brain concentration, probably
because of their inability to interact with cells [22].
The concentration of PEG-coated nanoparticles in
the central nervous system, especially in white
matter, was shown to be greatly increased in comparison to conventional non-PEG-coated nanoparticles. In addition, this increase was significantly
higher in pathological situations where blood–brain
barrier permeability is augmented and / or macro-
538
C. Vauthier et al. / Advanced Drug Delivery Reviews 55 (2003) 519–548
Fig. 11. Hypothetical mechanism of drug delivery to the brain by means of polysorbate 80-coated PACA nanospheres. (1) Adsorption of
apolipoprotein E (ApoE) onto the nanospheres; (2) transcytosis of the nanospheres; (3) endocytosis followed by intracellular degradation of
the nanospheres, resulting in release of the drug and diffusion towards the interior of the brain; (4) inhibition of P-glycoprotein; (5)
modulation of tight junction opening.
phages have infiltrated. Passive diffusion and macrophage uptake in inflammatory lesions seems to be the
mechanism underlying such brain penetration. This
was clearly documented after administration of PEGcoated nanospheres to rats bearing an experimental
allergic encephalomyelitis [145]. In addition, these
PEG-coated nanospheres showed, in comparison
with conventional non-PEG-coated nanoparticles, a
higher uptake by the brain of scrapie-infected animals, which may be useful for targetting drugs for
the treatment of prion diseases [65].
3.5.2. Oral route
There are numerous reports showing that uptake
and translocation of nanoparticles and microparticles
takes place after oral administration to animals [146–
148]. Different mechanisms have been proposed to
explain the translocation of particulate material
across the intestine: (i) uptake via Peyer’s patches or
isolated lymphoid follicles; (ii) intracellular uptake;
and (iii) intercellular / paracellular passage [149]. The
mechanism of nanoparticle uptake depends on the
nature of the nanoparticle. The uptake of PACA
nanocapsules by Peyer’s patches has been shown by
´
Damge et al. [148]. When administered in the lumen
of an isolated ileal segment of the rat, PACA
nanocapsules were found preferentially over Peyer’s
patches, through which they passed massively and
rapidly [148]. Nanocapsules were clearly visible in
M-cells and in intercellular spaces around the lymph
cells. Intracellular uptake of nanospheres has been
proposed by Kreuter et al. [150] based on electronmicroscopic autoradiographic investigations showing
radioactivity in epithelial and goblet cells after oral
administration of poly(hexylcyanoacrylate) nanospheres labeled with 14 C. The translocation of particles by a paracellular pathway was evidenced in a
study performed by Aprahamian et al. [151] using
poly(isobutylcyanoacrylate)
nanocapsules.
The
nanocapsules were filled with an iodinized oil
(lipiodol) in order to render them detectable using a
scanning electron microscope equipped with an
energy-dispersive X-ray spectrometer. When administered in an isolated segment of dog jejunum,
C. Vauthier et al. / Advanced Drug Delivery Reviews 55 (2003) 519–548
they appeared as vesicles associated with intraluminal mucus. Subsequently, they were observed in
intravillus capillaries in close contact with red blood
cells or adsorbed to the inner wall of endothelial
cells. Among these three mechanisms, and according
to many studies involving nanoparticles consisting of
other biodegradable and non-degradable polymers,
translocation via uptake in Peyer’s patches seems to
be the major pathway for rapid and substantial
passage after oral administration of nanoparticles.
Although it might occur in certain situations, the
passage of particles between absorptive cells is rather
less likely if the barrier of tight junctions has not
been disrupted. Although there are abundant reports
from various independent workers showing evidence
of the absorption of particulate systems by the
gastrointestinal tract, the oral absorption of
nanoparticles remains a controversial issue. However, even if a more exact estimation of the quantity
of absorbed particles is needed, as well as a better
understanding of the factors affecting particle uptake,
it must be concluded that translocation of small-sized
particles such as PACA nanoparticles is possible.
The question remains if the extent of particle translocation is compatible with a strategy of drug administration with therapeutic perspectives. This is discussed below.
3.5.2.1. Oral delivery of peptides, proteins and
vaccines
Poly(isobutylcyanoacrylate) nanocapsules were
shown 15 years ago to be able to encapsulate insulin
and to increase its activity as assessed by a reduction
of glycemia after oral feeding [56]. Several aspects
of this phenomenon are surprising: encapsulation of
a hydrophilic drug in the oily core of nanocapsules;
reduction of glycemia was only obtained with diabetic animals; and hypoglycemia appeared 2 days
after a single oral administration and was maintained
for up to 20 days depending on the insulin dose,
although the amplitude of the pharmacological effect
(minimum level of blood glucose) did not depend on
´
the insulin dose. Damge et al. [148] and Lowe and
Temple [58] suggested that nanocapsules could
protect insulin from proteolytic degradation in intestinal fluids as observed in the presence of different
proteolytic enzymes in vitro. Furthermore, later
539
studies showed that insulin did not react with the
alkylcyanoacrylate monomer during nanocapsule formation and was located within the oily core rather
than adsorbed on the surface [52].
The capacity of insulin nanocapsules to reduce
glycemia can also be explained by their translocation
´
through the intestinal barrier, as suggested by Damge
et al. [148]; for example, by the paracellular pathway
or via M-cells in Peyer’s patches [57]. Recently, the
use of Texas Red -labelled insulin allowed this
translocation to be visualized more readily [152].
One hour after oral administration, nanocapsules
reached the ileum. The presence of fluorescent areas
within the mucosa and even in the lamina propria
suggested that insulin-loaded PACA nanocapsules
could cross the intestinal epithelium. Although this
passage is certainly an important factor, it does not
explain the duration of the hypoglycemia. This
prolonged action could be due to the retention of a
proportion of the colloidal system in the gastrointestinal tract. Interestingly, a prolonged hypoglycemic effect was also observed with insulin
entrapped in PACA nanospheres when these nanospheres were dispersed in an oily phase containing a
surfactant [153]. This suggests that some components of the nanocapsules can act as absorption
promoters. Recently, insulin has also been encapsulated in water-containing nanocapsules [50]. These
nanocapsules, dispersed in a biocompatible microemulsion, could facilitate the intestinal absorption
of the encapsulated peptide after oral administration,
as suggested by the reduced blood glucose level
observed in diabetic rats [154].
Peptides other than insulin have been successfully
associated with nanocapsules. Among these, octreotide, a somatostatin analogue, has been incorporated in oil-containing PACA nanocapsules, and was
found to improve and prolong the therapeutic effect
of this peptide after administration by the oral route
[155]. Calcitonin has also been encapsulated in both
oil-containing and water-containing nanocapsules
[48,58,156].
Calcitonin-loaded,
oil-containing
nanocapsules showed behavior similar to insulinloaded, oil-containing nanocapsules [57,58]. Using
PACA nanocapsules with an aqueous core, the
effectiveness of the encapsulated peptide after oral
administration to rats was estimated as 45% of the
activity obtained after intravenous administration of
540
C. Vauthier et al. / Advanced Drug Delivery Reviews 55 (2003) 519–548
the same dose, whereas an absolute bioavailability of
40% was measured [48].
Even if the main limitation to the oral administration of PACA nanoparticles with peptides is that their
passage through the intestinal barrier is probably
restricted and sometimes erratic, they represent an
interesting tool for the oral delivery of antigens.
Indeed, M-cells appear to be the main site for the
uptake of PACA nanoparticles after oral administration [148] and, furthermore, it is generally accepted
that limited doses of antigen are sufficient for
mucous immunization. In fact, the oral delivery of
antigens may be considered the most convenient
means of producing an IgA antibody response.
However, this is limited by the enzymatic degradation of antigens in the GI tract and, additionally,
by their poor absorption. Thus, it has been postulated
that the use of micro- or nanoparticles for the oral
delivery of antigens can be efficient if these systems
are able to achieve protection of the antigenic
molecule. PACA nanoparticles have been shown to
enhance the secretory immune response after oral
administration in association with ovalbumin [157].
This result was not fully reproduced with poly(acrylamide) nanospheres loaded with the same
antigen, leading to the assumption that the antigen
was mainly located at the surface of the poly(acrylamide) nanospheres and could have been degraded during its passage through the gut. The
relatively high surface concentration of ovalbumin
adsorbed onto poly(butylcyanoacrylate) nanospheres
may have reduced the ability of the proteolytic
enzymes in the gut to gain access to, and to degrade,
the antigen, resulting in greater antigen availability.
3.5.2.2. Bioadhesive nanoparticles
Some polymers, either of natural or synthetic
origin, have the ability to adhere to wet mucosal
surfaces by means of hydrogen bonding or van der
Waals forces [158]. With swellable hydrophilic polymers, adhesion is optimal when mucosal contact is
made with the dry polymer. Further, progressive
hydration of the polymer leads to the formation of a
hydrogel, which is responsible for the development
of the considerable mucosa adhesion strength [158].
However, in the case of colloidal particles, bioadhesion is achieved with non-swellable polymers such as
PACA, and this is mainly due to the inherent
tendency of these small particles to develop intimate
contact on large mucosal sites [159]. The bioadhesive properties were found to vary with the size and
the surface characteristics of the nanoparticles [159–
161]. To improve the interactions of nanoparticles
with mucosae, lectins have been grafted onto the
nanoparticle surface [159], or nanoparticles have
been coated with chitosan [45].
Bioadhesion has been tested in vivo. After peroral
administration
of
radiolabelled
poly(hexylcyanoacrylate) nanoparticles to mice, whole-body
autoradiography showed that, 30 min after administration, the particles were exclusively localized in the
stomach [162]. After 4 h, a large quantity of
radioactivity was found in the intestine in the form of
clusters without macroradiographic evidence of accumulation at specific intestinal sites. On the contrary, a persistent film of nanoparticles adhering to
the stomach wall was observed. In this study, very
little of the radioactivity was found to be absorbed.
In a similar study, microautoradiographs confirmed
the presence of radioactivity throughout the gut
[150,162]. The amount of radioactivity decreased to
30–40% of the 90-min value within 4–8 h and to
5%, 24 h after dosing. Histological investigation
showed radioactivity adjacent to the brush border,
incorporated into the underlying cell layers and in
goblet cells up to 6 days after administration. However, the exclusive use of a radioactive tracer in
these experiments makes the presence of physically
intact particles 6 days after administration questionable because of possible degradation of the
particles in the gastrointestinal tract.
The pharmacokinetics of several drugs have been
improved after oral administration by means of
nanoparticles. Most studies were carried out with
conventional formulations, which means that the
carriers were not specifically designed to improve the
bioadhesion performance of the particles. The bioavailability of vincamine was about 25% when
administered in an aqueous solution to rabbits. After
oral administration of vincamine adsorbed on poly(hexylcyanoacrylate) nanoparticles, the bioavailability reached 40%, probably due to the prolonged
period of contact of the drug delivery system with
the mucosae [163]. Nanocapsules of poly(isobutylcyanoacrylate) increased the bioavailability
of iodine after administration of lipiodol, an iodized
C. Vauthier et al. / Advanced Drug Delivery Reviews 55 (2003) 519–548
oil, to the jejunum of dogs [164]. With the nanocapsules, the blood level of iodine was prolonged from
75 to over 105 min. This observation was also
attributed to the prolonged period of contact between
the lipiodol drug and the mucus of the microvilli
membrane [164]. Darodipine, a calcium flux inhibitor which causes strong vasodilatation, is characterized by a short half-life time ranging from 2 to 4 h.
The use of nanocapsules resulted in a reduction in
the intensity of the initial very strong hypotensive
effect and prolonged the pharmacological activity of
the drug [165].
3.5.2.3. Application to the administration of antiproteases
Saquinavir is a potent HIV-1 and HIV-2 protease
inhibitor that has been approved for use in the
treatment of patients with acquired immunodeficiency syndrome. However, administering
saquinavir by the oral route is a formidable challenge
due to its poor absorption pattern. Thus several
approaches were investigated to improve its oral
bioavailability, among them its association with
PACA nanoparticles. Saquinavir-loaded PACA
nanospheres could be easily prepared in the presence
of a drug–cyclodextrin complex. It was found that
large amounts of cyclodextrins remained associated
with the particles, resulting in a 20-fold increase in
saquinavir loading compared to nanoparticles prepared in the absence of cyclodextrins [36]. This
study showed that the loading of saquinavir in PACA
nanospheres could be improved dramatically by
simultaneously increasing the apparent solubility of
the drug in the preparation medium and the amount
of cyclodextrin associated with the particles, making
these nanospheres an interesting system for oral
application. Indeed, Boudad et al. [166] have shown
that this system is able to improve significantly the
amount and kinetics of saquinavir transported from
apical to basolateral sites in the CACO-2 monolayer
cell model.
3.5.3. Other routes of administration
PACA nanoparticles have been evaluated as controlled release devices for peptide delivery after
subcutaneous administration. Radiolabelled poly(isobutylcyanoacrylate) nanospheres were injected
subcutaneously and the autoradiographic pictures
541
revealed a progressive reduction of staining in
muscular tissue, suggesting that the nanospheres
were progressively biodegraded [167]. These nanospheres were found to release growth releasing factor
in a sustained manner and to improve its bioavailability [39]. After association with the nanospheres,
the peptide was partially protected from enzymatic
degradation, whereas it was very rapidly metabolized
at the injection site when administered as the free
drug.
Nanoparticle technologies have also been investigated for the administration of drugs to the eye.
However, in preliminary experiments, PACA
nanoparticles appeared not to be very well tolerated
by the ocular mucosae, leading to cell lysis [168].
Thus, the development of ocular therapy by means of
nanoparticles has considered the use of other polymers, among which poly(´-caprolactone) showed
good potential for eye administration. Very recently,
progress in the area of the research and development
of new formulations for ocular therapy encouraged
some authors to reconsider PACA nanoparticles in
opthalmology. To reduce the observed toxic effect to
the eye, PACA nanoparticles were either dispersed in
a poly(acrylic acid) or PEG gel [169,170] or coated
with PEG chains [171]. These approaches were
shown to improve the tolerance of the ocular
mucosae to PACA nanoparticles and open new
avenues for the further development of such formulations as drug delivery systems for ocular therapy.
4. Conclusion
There are now a significant number of technologies based on cyanoacrylate monomers or polymers for biomedical applications. This is due to the
fact that cyanoacrylate monomers are able to form
polymeric materials with biodegradable characteristics which may easily be controlled depending on the
nature of the cyanoacrylic monomer used. On the
other hand, it is unquestionable that the use of
cyanoacrylates as surgical glue represents, from a
toxicological point of view, a favorable situation.
Among the different cyanoacrylate-based technologies, nanoparticles probably offer the most exciting possibilities in terms of medical applications.
Indeed, they allow modification of the intracellular
542
C. Vauthier et al. / Advanced Drug Delivery Reviews 55 (2003) 519–548
trafficking of drugs, thus opening new ways to reach,
in a controlled manner, the intracellular bacteria
responsible for opportunistic diseases or to administer anticancer compounds to cells in a manner that is
able to bypass the cell P-glycoprotein detoxification
process. Also, there are interesting perspectives for
the intracellular delivery of molecules such as oligonucleotides, which are able, when protected from
nucleases, to modulate gene expression. However, as
stated above, many questions remain concerning the
intracellular fate of nanoparticles, probably because
the answers differ from one cell line to another and
from one cyanoacrylic polymer to another. Whatever
the answer, there is an urgent need to design
nanoparticles able to specifically deliver these molecules, either to the cytoplasm or to the nucleus
depending on the target. It is evident that, due to the
drawbacks of viral vectors, there is a challenge to
deliver genetic material intracellularly by means of
nanodispersed synthetic carriers and this represents a
new challenge for cyanoacrylate polymers.
Controlling the tissue distribution of nanoparticles
after intravascular administration is another challenge, which has been partly solved by the newly
designed poly(PEGCA-co-PACA) copolymer that
avoids opsonisation, thus reducing liver and spleen
uptake and increasing the circulation time in the
blood. Due to the leaky vasculature in numerous
brain pathologies, very exciting applications are now
envisioned in this field. Other, very recent approaches allow the efficient coating of PACA
nanoparticles with different polysaccharides, which
are also expected to modify the tissue distribution of
these particles. However, the exciting potential of
this approach has not yet been completely investigated.
The oral administration of peptides and proteins
by means of PACA nanoparticles is another interesting perspective. Although the oral absorption of
nanoparticles remains a controversial field of research, it would be inconsistent to completely ignore
this approach since interesting results have been
obtained by various independent research groups.
Thus, as discussed above, nanoparticles could also
open up very interesting perspectives for the oral
delivery of antigens.
Finally, it is surprising that there has been so very
little effort expended to develop new molecularly
addressed PACA nanoparticles. Indeed, the targeting
of these nanotechnologies needs to be urgently
improved by adequate decoration of the nanoparticle
surface. In this respect, the preliminary results
obtained with folic acid for targeting of the folic acid
receptor are encouraging, but deserve further development.
The concept of cyanoacrylate polymers for the
design of new drug delivery systems emerged from
academic pharmaceutical research in Europe. In the
last 15 years, cyanoacrylate-based technology has
improved and its potential in therapeutics is well
documented. Cyanoacrylate nanoparticles have recently entered Phase II clinical trials and results are
expected in the treatment of resistant cancers.
References
[1] P. Couvreur, B. Kante, M. Roland, P. Guiot, P. Bauduin, P.
Speiser, Polycyanoacrylate nanocapsules as potential
lysosomotropic carriers: preparation, morphological and
sorptive properties, J. Pharm. Pharmacol. 31 (1979) 331–
332.
[2] F. Leonard, R.K. Kulkarni, G. Brandes, J. Nelson, J.J.
Cameron, Synthesis and degradation of poly(alkyl acyanoacrylates), J. Appl. Polym. Sci. 10 (1996) 259–272.
[3] I. Skeist, J. Miron, Adhesive composition, in: N. Bikales
(Ed.), Encyclopedia of Polymer Science and Engineering,
Suppl. Vol. 2, Wiley–Interscience, New York, 1977, pp.
1–19.
[4] H.W. Coover, D.W. Dreifus, J.T. O’Connor, Cyanoacrylate
adhesives, in: I. Skeist (Ed.), Handbook of Adhesives, 3rd
Edition, Van Nostrand Reinhold, New York, 1990, pp. 463–
477.
[5] M.E. King, A.Y. Kinney, Tissue adhesives: a new method of
wound repair, Nurse Pract. 24 (1999) 66–73.
[6] H. Oowaki, S. Matsuda, N. Sakai, T. Ohta, H. Iwata, A.
Sadato, W. Taki, N. Hashimoto, Y. Ikada, Non-adhesive
cyanoacrylate as an embolic material for endovascular
neurosurgery, Biomaterials 21 (2000) 1039–1046.
[7] T.B. Reece, T.S. Maxey, I.L. Kron, A prospectus on tissue
adhesives, Am. J. Surg. 182 (2001) 40S–44S.
[8] J.S. Pollak, R.I. White Jr., The use of cyanoacrylate adhesives in peripheral embolization, J. Vasc. Interv. Radiol. 12
(2001) 907–913.
[9] F. De Jaeghere, E. Doelker, R. Gurny, Nanoparticles, in: E.
Mathiowitz (Ed.), The Encyclopedia of Controlled Drug
Delivery, Wiley, New York, 1999, pp. 641–664.
[10] N.M.V.R. Kumar, Nano and microparticles as controlled drug
delivery devices, J. Pharm. Pharm. Sci. 3 (2000) 234–258.
[11] K.S. Soppimath, T.M. Aminabhavi, A.R. Kulkarni, W.E.
C. Vauthier et al. / Advanced Drug Delivery Reviews 55 (2003) 519–548
[12]
[13]
[14]
[15]
[16]
[17]
[18]
[19]
[20]
[21]
[22]
[23]
[24]
[25]
Rudzinski, Biodegradable polymeric nanoparticles as drug
delivery devices, J. Control. Release 70 (2001) 1–20.
P. Couvreur, G. Barratt, E. Fattal, P. Legrand, C. Vauthier,
Nanocapsule technology: a review, Crit. Rev. Drug Deliv.
Syst. 19 (2002) 99–134.
E. Fattal, C. Vauthier, Nanoparticles as drug delivery systems, in: J. Swarbrick, J.C. Boylan (Eds.), Encyclopedia of
Pharmaceutical Technology, Marcel Dekker, New York,
2002, pp. 1874–1892.
C. Vauthier, P. Couvreur, Degradation of poly(alkylcyanoacrylates), in: J.P. Matsumara, A. Steinbuchel
(Eds.), Miscellaneous Biopolymers and Biodegradation of
Synthetic Polymers, Handbook of Biopolymers, Vol. 9,
Wiley–VHC, New York, 2002, in press.
I. Brigger, C. Dubernet, P. Couvreur, Nanoparticles in cancer
therapy and diagnosis, Adv. Drug Deliv. Rev. (in press).
P. Couvreur, B. Kante, M. Roland, Les vecteurs lysosomotropes, J. Pharm. Belg. 35 (1980) 51–60.
P. Couvreur, B. Kante, L. Grislain, M. Roland, P. Speiser,
Toxicity of polyalkylcyanoacrylate nanoparticles. II. Doxorubicin-loaded nanoparticles, J. Pharm. Sci. 71 (1982) 790–
792.
G. Barratt, G. Couarraze, P. Couvreur, C. Dubernet, E. Fattal,
R. Gref, D. Labarre, P. Legrand, G. Ponchel, C. Vauthier,
Polymeric micro and nanoparticles as drug carriers, in: S.
Dumitriu (Ed.), Polymeric Biomaterials, 2nd Edition, Marcel
Dekker, New York, 2001, pp. 753–782.
J. Kattan, J.P. Droz, P. Couvreur, J.P. Marino, A. BoutanLaroze, P. Rougier, P. Brault, H. Vranks, J.M. Grognet, X.
Morge, H. Sancho-Garnier, Phase I clinical trial and pharmacokinetic evaluation of doxorubicin carried by polyisohexylcyanoacrylate nanoparticles, Invest. New Drugs 10
(1992) 191–199.
¨
B. Stella, S. Arpicco, M.T. Peracchia, D. Desmaele, J.
Hoebeke, M. Renoir, J. D’Angelo, L. Cattel, P. Couvreur,
Design of folic acid-conjugated nanoparticles for drug
targeting, J. Pharm. Sci. 89 (2000) 1452–1464.
I. Brigger, P. Chaminade, V. Marsaud, M. Appel, M. Besnard,
R. Gurny, M. Renoir, P. Couvreur, Tamoxifen encapsulation
within polyethylene glycol-coated nanospheres. A new antiestrogen formulation, Int. J. Pharm. 214 (2001) 37–42.
P. Calvo, B. Gouritin, H. Chacun, D. Desmaele, J. D’Angelo,
J.P. Noel, D. Georgin, E. Fattal, J.P. Andreux, P. Couvreur,
Long-circulating PEGylated polycyanoacrylate nanoparticles
as new drug carrier for brain delivery, Pharm. Res. 18 (2001)
1157–1166.
Y.P. Li, Y.Y. Pei, Z.H. Zhou, X.Y. Zhang, Z.H. Gu, J. Ding,
J.J. Zhou, X.J. Gao, J.H. Zhu, Stealth polycyanoacrylate
nanoparticles as tumor necrosis factor-a carriers: pharmacokinetics and anti-tumor effects, Biol. Pharm. Bull. 24
(2001) 662–665.
D.C. Pepper, Anionic and zwitterionic polymerization of
a-cyanoacrylates, J. Polym. Sci. Polym. Symp. 62 (1978)
65–77.
B. Ryan, G. McCann, Novel sub-ceiling temperature rapid
depolymerization–repolymerization
reactions
of
cyanoacrylate polymers, Macromol. Rapid Commun. 17
(1996) 217–227.
543
[26] G.G. Hollock, Expanded applications for octyl-2cyanoacrylate as a tissue adhesive, Ann. Plast. Surg. 47
(2001) 576.
[27] R. Marcovich, A.L. Williams, M.A. Rubin, J.S. Wolf, Comparison of 2-octyl cyanoacrylate adhesive, fibrin glue, and
suturing for wound closure in the porcine urinary tract,
Urology 57 (2001) 806–810.
[28] A. Kitoh, Y. Arima, C. Nishigori, K. Kikuta, Y. Miyachi,
Tissue adhesive and postoperative allergic meningitis, Lancet
359 (2002) 1669–1670.
[29] D.T. Burns, J.K. Brown, A. Dinsmore, K. Harvey, Baseactivated latent fingerprints fumed with a cyanoacrylate
monomer. A quantitative study using Fourrier-transformed
infra-red spectroscopy, Anal. Chim. Acta 362 (1998) 171–
176.
[30] A.J. Singer, J.V. Quinn, R.E. Clark, J.E. Hollander, Closure
of lacerations and incision with octylcyanoacrylate: a multicenter randomized controlled trial, Surgery 131 (2002)
270–276.
[31] D.K. Bahsin, N.J. Malhi, Variceal bleeding and portal hypertension: much to learn, much to explore, Endoscopy 34
(2002) 119–128.
[32] F. Lescure, C. Zimmer, D. Roy, P. Couvreur, Optimization of
polycyanoacrylate nanoparticle preparation: influence of
sulfur dioxide and pH on nanoparticle characteristics, J.
Colloid Interface Sci. 154 (1992) 77–86.
[33] S.J. Douglas, L. Illum, S.S. Davis, Particle size and size
distribution of poly(butyl 2-cyanoacrylate) nanoparticles. II.
Influence of stabilizers, J. Colloid Interface Sci. 103 (1985)
154–163.
[34] M.J. Alonso, A. Sanchez, D. Torres, B. Seijo, J.L. Vila-Jato,
Joint effect of monomer and stabilizer concentrations on the
physico-chemical
characteristics
of
poly(butyl-2cyanoacrylate) nanoparticles, J. Microencapsul. 7 (1990)
517–526.
[35] B. Seijo, E. Fattal, L. Roblot-Treupel, P. Couvreur, Design of
nanoparticles of less than 50 nm diameter: preparation,
characterization and drug loading, Int. J. Pharm. 62 (1990)
1–7.
ˆ
[36] H. Boudad, P. Legrand, G. Lebas, M. Cheron, D. Duchene,
G.G. Ponchel, Combined hydroxypropyl-beta-cyclodextrin
and poly(alkylcyanoacrylate) nanoparticles intended for oral
administration of saquinavir, Int. J. Pharm. 218 (2001) 113–
124.
[37] M.M. Gallardo, L. Roblot-Treupel, J. Mahuteau, I. Genin, P.
Couvreur, M. Plat, F. Puisieux, Nanocapsules et nanospheres
`
d’alkylcyanoacrylate. Interaction principe actif-polymere, in:
Proceedings of the 5th International Congress on Pharmaceutical Technology, 1989, pp. 36–45, Vol. 4.
[38] V. Guise, J.Y. Drouin, J. Benoit, J. Mahuteau, P. Dumont, P.
Couvreur, Vidarabine-loaded nanoparticles: a physicochemical study, Pharm. Res. 7 (1990) 736–741.
[39] J.L. Grangier, M. Puygrenier, J.C. Gautier, P. Couvreur,
Nanoparticles as carriers for growth hormone releasing
factor, J. Control. Release 15 (1991) 3–13.
[40] A. Labib, V. Lenaerts, F. Chouinard, J.C. Leroux, R. Ouellet,
J.E. Van Lier, Biodegradable nanospheres containing phthalo-
544
[41]
[42]
[43]
[44]
[45]
[46]
[47]
[48]
[49]
[50]
[51]
[52]
[53]
[54]
C. Vauthier et al. / Advanced Drug Delivery Reviews 55 (2003) 519–548
cyanines and naphthalocyanines for targeted photodynamic
tumor therapy, Pharm. Res. 8 (1991) 1027–1031.
G.E. Ghanem, C. Joubran, R. Arnould, F. Lejeune, J.
Fruhling, Labelled polycyanoacrylate nanoparticles for
human in vivo use, Appl. Radiat. Isot. 44 (1993) 1219–1224.
S.J. Douglas, L. Illum, S.S. Davis, Poly(butyl-2cyanoacrylate) nanoparticles with differing surface charges,
J. Control. Release 3 (1986) 15–23.
M.T. Peracchia, C. Vauthier, F. Puisieux, P. Couvreur,
Development of sterically stabilized poly(isobutyl 2cyanoacrylate) nanoparticles by chemical coupling of poly(ethylene glycol), J. Biomed. Mater. Res. 34 (1997) 317–
326.
M.T. Peracchia, C. Vauthier, M.I. Popa, F. Puisieux, P.
Couvreur, Investigation of the formation of sterically stabilized poly(ethylene glycol / isobutylcyanoacrylate) nanoparticles by chemical grafting of polyethylene glycol during the
polymerization of isobutyl cyanoacrylate, S.T.P. Pharma Sci.
7 (1997) 513–520.
S.C. Yang, H.X. Ge, Y. Hu, X.Q. Jiang, C.Z. Yang, Formation
of positively charged poly(butyl cyanoacrylate) nanoparticles
stabilized by chitosan, Coll. Polym. Sci. 278 (2000) 285–
292.
N. Al Khoury-Fallouh, L. Roblot-Treupel, H. Fessi, J.P.
Devissaguet, F. Puisieux, Development of a new process for
the manufacture of poly(isobutylcyanoacrylate) nanocapsules, Int. J. Pharm. 28 (1986) 125–136.
M. Gasco, M. Trotta, Nanoparticles from microemulsions,
Int. J. Pharm. 29 (1986) 267–268.
H. Vranckx, M. Demoustier, M. Deleers, A new nanocapsule
formulation with hydrophilic core: application to the oral
administration of salmon calcitonin in rats, Eur. J. Pharm.
Biopharm. 42 (1996) 345–347.
G. Lambert, E. Fattal, H. Pinto-Alphandary, A. Gulik, P.
Couvreur, Polyisobutylcyanoacrylate nanocapsules containing an aqueous core as a novel colloidal carrier for the
delivery of oligonucleotides, Pharm. Res. 17 (2000) 707–
714.
S. Watnasirichaikul, N.M. Davies, R. Rades, I.G. Tucker,
Preparation of biodegradable insulin nanocapsules from
biocompatible microemulsions, Pharm. Res. 17 (2000) 684–
689.
M.M. Gallardo, G. Couarraze, B. Denizot, L. Treupel, P.
Couvreur, F. Puisieux, Preparation and purification of isohexylcyanoacrylate nanocapsules, Int. J. Pharm. 100 (1993)
55–64.
M. Aboubakar, F. Puisieux, P. Couvreur, M. Deyme, C.
Vauthier, Study of the mechanism of insulin encapsulation in
poly(isobutylcyanoacrylate) nanocapsules obtained by interfacial polymerization, J. Biomed. Mater. Res. 47 (1999)
568–576.
¨
M. Wohlgemuth, W. Machtle, C. Mayer, Improved preparation and physical studies of polybutylcyanoacrylate nanocapsules, J. Microencapsul. 17 (2000) 437–448.
G. Puglisi, M. Fresta, G. Giammona, C.A. Venture, Influence
of the preparation conditions in poly(ethylcyanoacrylate)
nanocapsules formation, Int. J. Pharm. 125 (1995) 283–287.
[55] M. Fresta, G. Cavallaro, G. Giammona, E. Wehrli, G. Puglisi,
Preparation
and
characterization
of
polyethyl-2cyanoacrylate nanocapsules containing antiepileptic drugs,
Biomaterials 17 (1996) 751–758.
[56] C. Damge, C. Michel, M. Aprahamian, P. Couvreur, New
approach for oral administration of insulin with polyalkylcyanoacrylate nanocapsules as drug carrier, Diabetes 37
(1988) 246–251.
[57] C. Michel, M. Aprahamian, L. Defontaine, P. Couvreur, C.
Damge, The effect of site of administration in the gastrointestinal tract on the absorption of insulin from nanocapsules
in diabetic rats, J. Pharm. Pharmacol. 43 (1990) 1–5.
[58] P.J. Lowe, C.S. Temple, Calcitonin and insulin in isobutylcyanoacrylate nanocapsules: protection against proteases and effect on intestinal absorption in rats, J. Pharm.
Pharmacol. 46 (1994) 547–552.
[59] Y.K. Choi, Y.H. Kim, S.W. Kim, Block copolymer nanoparticles of ethylene oxide and isobutyl cyanoacrylate, Macromolecules 28 (1995) 8419–8421.
¨
[60] M.T. Peracchia, D. Desmaele, P. Couvreur, J. D’Angelo,
Synthesis of a novel poly(MePEG cyanoacrylate-coalkylcyanoacrylate) amphiphilic copolymer for nanoparticle
technology, Macromolecules 30 (1997) 846–851.
¨
[61] M.T. Peracchia, D. Desmaele, C. Vauthier, D. Labarre, E.
Fattal, J. D’Angelo, P. Couvreur, Development of novel
technologies for the synthesis of biodegradable pegylated
nanoparticles, in: G. Gregoriadis, B. McCormack (Eds.),
Strategies for Stealth Therapeutic Systems, Targeting of
Drugs, Vol. 6, Plenum Press, New York, 1998, pp. 225–239.
¨
[62] M.T. Peracchia, C. Vauthier, D. Desmaele, A. Gulik, J.C.
Dedieu, M. Demoy, J. D’Angelo, P. Couvreur, Pegylated
nanoparticles from a novel methoxypolyethylene glycol
cyanoacrylate–hexadecyl cyanoacrylate amphiphilic copolymer, Pharm. Res. 15 (1998) 548–554.
[63] M.T. Peracchia, E. Fattal, D. Desmaele, M. Besnard, J.P.
Noel, J.M. Gomis, M. Appel, J. d’Angelo, P. Couvreur,
Stealth PEGylated polycyanoacrylate nanoparticles for intravenous administration and splenic targeting, J. Control.
Release 60 (1999) 121–128.
[64] Y.P. Li, Y.Y. Pei, Z.H. Zhou, X.Y. Zhang, Z.H. Gu, J. Ding,
J.J. Zhou, X.J. Gao, PEGylated polycyanoacrylate nanoparticles as tumor necrosis factor-a carrier, J. Control. Release 71
(2001) 287–296.
[65] P. Calvo, B. Gouritin, I. Brigger, C. Lasmezas, J. Deslys, A.
Williams, J.P. Andreux, D. Dormont, P. Couvreur, PEGylated
polycyanoacrylate nanoparticles as vector for drug delivery
in prion diseases, J. Neurosci. Methods 111 (2001) 151–155.
[66] V. Lenaerts, P. Couvreur, D. Christiaens-Leyh, E. Joiris, M.
Roland, B. Rollman, P. Speiser, Degradation of poly(isobutyl
cyanoacrylate) nanoparticles, Biomaterials 5 (1984) 65–68.
[67] L. Vansnick, P. Couvreur, D. Christiaens-Ley, M. Roland,
Molecular weights of free and drug-loaded nanoparticles,
Pharm. Res. 1 (1985) 36–41.
¨
[68] K. Langer, E. Seegmuller, A. Zimmer, J. Kreuter, Characterization of polybutylcyanoacrylate nanoparticles: quantification of PBCA polymer and dextran, Int. J. Pharm. 110
(1994) 21–27.
C. Vauthier et al. / Advanced Drug Delivery Reviews 55 (2003) 519–548
¨
[69] R.H. Muller, C. Lherm, J. Herbort, P. Couvreur, In vitro
model for the degradation of alkylcyanoacrylate nanoparticles, Biomaterials 11 (1990) 590–595.
[70] D. Scherer, J.R. Robinson, J. Kreuter, Influence of enzymes
on the stability of polybutylcyanoacrylate nanoparticles, Int.
J. Pharm. 101 (1994) 165–168.
[71] C. Lherm, R. Muller, F. Puisieux, P. Couvreur, II. Cytotoxicity of cyanoacrylate nanoparticles with different alkyl chain
length, Int. J. Pharm. 84 (1992) 13–22.
[72] C.W. Wade, F. Leonard, Degradation of poly(methylcyanoacrylates), J. Biomed. Mater. Res. 6 (1972)
215–220.
[73] W.R. Vezin, A.T. Florence, In vitro heterogenous degradation
of poly(n-alkyl a-cyanoacrylates), J. Biomed. Mater. Res. 14
(1980) 93–106.
[74] H.V. Vinters, H.W. Ho, Effects of isobutyl-2-cyanoacrylate
polymer on cultured cells derived from murine cerebral
microvessels, Toxicol. In Vitro 2 (1988) 37–41.
[75] Y.C. Tseng, S.H. Hyon, Y. Ikada, Modification of the
synthesis and investigation of properties for 2cyanoacrylates, Biomaterials 11 (1990) 73–79.
[76] A.W. Cooper, P.J. Harris, G.K. Kumar, J.C. Tebby, Hydrolysis
of
alkylcyanoacrylate
and
ethoxycarbonylpropenoylphosphonate polymers, J. Polym. Sci. Part A:
Polym. Chem. 27 (1989) 1967–1974.
[77] D. Ley, P. Couvreur, V. Lenaerts, M. Roland, P. Speiser,
´
´
Etude du mecanisme de degradation des nanoparticules de
polycyanoacrylate d’alkyle, Labo Pharma-Probl. Tech. 32
(1984) 100–104.
[78] L. Grislain, P. Couvreur, V. Lenaerts, M. Roland, D. DeprezDecampeneere, P. Speiser, Pharmacokinetics and distribution
of a biodegradable drug-carrier, Int. J. Pharm. 15 (1983)
335–345.
[79] V. Lenaerts, J.F. Nagelkerke, T.J. Van Berkel, P. Couvreur, L.
Grislain, M. Roland, P. Speiser, In vivo uptake of poly¨
isobutyl cyanoacrylate nanoparticles by rat liver Kupffer,
endothelial, and parenchymal cells, J. Pharm. Sci. 73 (1984)
980–982.
[80] S. Gibaud, J.P. Andreux, C. Weingarten, M. Renard, P.
Couvreur, Increased bone marrow toxicity of doxorubicin
bound to nanoparticles, Eur. J. Cancer 6 (1994) 820–826.
[81] M. Demoy, S. Gibaud, J.P. Andreux, C. Weingarten, B.
Gouritin, P. Couvreur, Splenic trapping of nanoparticles:
complementary approaches for in situ studies, Pharm. Res.
14 (1997) 463–468.
[82] C. Chavany, T. Ledoan, P. Couvreur, F. Puisieux, C. Helene,
Polyalkylcyanoacrylate nanoparticles as polymeric carriers
for antisense oligonucleotides, Pharm. Res. 9 (1992) 441–
449.
[83] R. Gref, Y. Minamitake, M.T. Peracchia, V. Trubetskoy, V.
Torchilin, R. Langer, Biodegradable long-circulating polymeric nanospheres, Science 263 (1994) 1600–1603.
[84] D. Bazile, C. Prud’homme, M.T. Bassoullet, M. Marlard, G.
Spenlehauer, M. Veillard, Stealth Me.PEG-PLA nanoparticles
avoid uptake by the mononuclear phagocytes system, J.
Pharm. Sci. 84 (1995) 493–498.
[85] C. Verdun, F. Brasseur, H. Vranckx, P. Couvreur, M. Roland,
[86]
[87]
[88]
[89]
[90]
[91]
[92]
[93]
[94]
[95]
[96]
[97]
[98]
[99]
545
Tissue distribution of doxorubicin associated with polyisohexylcyanoacrylate nanoparticles, Cancer Chemother.
Pharmacol. 26 (1990) 13–18.
N. Chiannilkulchai, N. Ammoury, B. Caillou, J.P. Devissaguet, P. Couvreur, Hepatic tissue distribution of doxorubicin-loaded nanoparticles after i.v. administration in reticulosarcoma M 5076 metastasis-bearing mice, Cancer
Chemother. Pharmacol. 26 (1990) 122–126.
F. Brasseur, C. Verdun, P. Couvreur, C. Deckers, M. Roland,
´
´ ´
Evaluation experimentale de l’efficacite therapeutique de la
´
´
doxorubicine associee associee aux nanoparticules de polyalkylcyanoacrylate, in: Proceeding of the 4th International
Conference on Pharmaceutical Technology, Paris, 1986.
R. Fernandez-Urrusuno, E. Fattal, J.M. Rodrigues Jr., J.
Feger, P. Bedossa, P. Couvreur, Effect of polymeric
nanoparticle administration on the clearance activity of the
mononuclear phagocyte system in mice, J. Biomed. Mater.
Res. 31 (1996) 401–408.
N. Ammoury, H. Fessi, J.P. Devissaguet, F. Puisieux, S.
Benita, Physicochemical characterization of polymeric
nanocapsules and in vitro release of indomethacin as a drug
model, S.T.P. Pharma 5 (1989) 642–646.
´
P. Couvreur, H. Vranckx, F. Brasseur, M. Roland, Toxicite
`
des nanoparticles a base de polycyanoacrylates d’alkyle,
S.T.P. Pharma Sci. 5 (1989) 31–37.
S. Maassen, E. Fattal, R. Muller, P. Couvreur, Cell cultures
for the assessment of toxicity and uptake of polymeric drug
carriers, S.T.P. Pharma Sci. 3 (1993) 11–22.
B. Kante, P. Couvreur, G. Dubois-Krack, C. De Meester, P.
Guiot, M. Roland, M. Mercier, P. Speiser, Toxicity of
polyalkylcyanoacrylate nanoparticles. I. Free nanoparticles,
J. Pharm. Sci. 71 (1982) 786–790.
J. Kreuter, C.G. Wilson, J.R. Fry, P. Paterson, J.H. Ratcliffe,
Toxicity and association of polycyanoacrylate nanoparticles
with hepatocytes, J. Microencapsul. 1 (1984) 253–257.
C. Kubiak, P. Couvreur, L. Manil, B. Clausse, Increased
cytotoxicity of nanoparticle-carried adriamycin in vitro and
potentiation by verapamil and amiodarone, Biomaterials 10
(1989) 553–556.
E. Gipps, P. Groscurt, J. Kreuter, P.P. Speiser, The effects of
poly(alkylcynoacrylate) nanoparticles on human normal and
malignant mesenchymal cells in vitro, Int. J. Pharm. 40
(1987) 23–31.
R. Gaspar, V. Preat, F.R. Opperdoes, M. Roland, Macrophage
activation
by
polymeric
nanoparticles
of
polyalkylcyanoacrylates: activity against intracellular Leishmania
donovani associated with hydrogen peroxide production,
Pharm. Res. 9 (1992) 782–787.
R. Fernandez-Urrusuno, E. Fattal, D. Porquet, J. Feger, P.
Couvreur, Evaluation of liver toxicological effects induced
by polyalkylcyanoacrylate nanoparticles, Toxicol. Appl.
Pharmacol. 130 (1995) 272–279.
´
R. Fernandez-Urrusuno, E. Fattal, J. Feger, P. Couvreur, P.
´
Theron, Evaluation of hepatic antioxidant systems after
intravenous administration of polymeric nanoparticles,
Biomaterials 18 (1997) 511–517.
P. Couvreur, P. Tulkens, M. Roland, A. Trouet, P. Speiser,
546
C. Vauthier et al. / Advanced Drug Delivery Reviews 55 (2003) 519–548
Nanocapsules: a new lysosomotropic carrier, FEBS Lett. 84
(1977) 323–326.
[100] V. Guise, P. Jaffray, J. Delattre, F. Puisieux, M. Adolphe, P.
Couvreur, Comparative cell uptake of propidium iodide
associated with liposomes or nanoparticles, Cell. Mol. Biol.
33 (1987) 397–405.
[101] E. Fattal, M. Youssef, P. Couvreur, A. Andremont, Treatment of experimental salmonellosis in mice with ampicillinbound nanoparticles, Antimicrob. Agents Chemother. 33
(1989) 1540–1543.
[102] M. Youssef, E. Fattal, M.J. Alonso, L. Roblot-Treupel, J.
Sauzieres, C. Tancrede, A. Omnes, P. Couvreur, A. Andremont, Effectiveness of nanoparticle-bound ampicillin in
the treatment of Listeria monocytogenes infection in
athymic nude mice, Antimicrob. Agents Chemother. 32
(1988) 1204–1207.
[103] F. Forestier, P. Gerrier, C. Chaumard, A.M. Quero, P.
Couvreur, C. Labarre, Effect of nanoparticle-bound ampicillin on the survival of Listeria monocytogenes in mouse
peritoneal macrophages, J. Antimicrob. Chemother. 30
(1992) 173–179.
[104] O. Balland, H. Pinto-Alphandary, S. Pecquet, A. Andremont, P. Couvreur, The uptake of ampicillin-loaded
nanoparticles by murine macrophages infected with Salmonella typhimurium, J. Antimicrob. Chemother. 33 (1994)
509–522.
[105] H. Pinto-Alphandary, O. Balland, M. Laurent, A. Andremont, F. Puisieux, P. Couvreur, Intracellular visualization of
ampicillin-loaded nanoparticles in peritoneal macrophages
infected in vitro with Salmonella typhimurium, Pharm. Res.
11 (1994) 38–46.
[106] N.A. Buchmeier, F. Heffron, Inhibition of macrophage
phagosome–lysosome fusion by Salmonella typhimurium,
Infect. Immun. 59 (1991) 2232–2238.
[107] M.E. Page-Clisson, H. Pinto-Alphandary, E. Chachaty, P.
Couvreur, A. Andremont, Drug targeting by polyalkylcyanoacrylate nanoparticles is not efficient against
persistent Salmonella, Pharm. Res. 15 (1998) 544–549.
[108] R. Gaspar, F.R. Opperdoes, V. Preat, M. Roland, Drug
targeting with polyalkylcyanoacrylate nanoparticles—in
vitro activity of primaquine-loaded nanoparticles against
intracellular Leishmania donovani, Ann. Trop. Med.
Parasitol. 86 (1992) 782–787.
[109] M. Fouarge, M. Dewulf, P. Couvreur, M. Roland, H.
Vranckx, Development of dehydroemetine nanoparticles for
the treatment of visceral leishmaniasis, J. Microencapsul. 6
(1989) 29–34.
[110] N. Chiannilkulchai, Z. Driouich, J.P. Benoit, A.L. Parodi, P.
Couvreur, Doxorubicin-loaded nanoparticles: increased efficiency in murine hepatic metastases, Sel. Cancer Ther. 5
(1989) 1–11.
[111] P. Couvreur, L. Grislain, V. Lenaerts, F. Brasseur, P. Guiot,
A. Biornacki, Biodegradable polymeric nanoparticles as
drug carrier for antitumor agents, in: P. Guiot, P. Couvreur
(Eds.), Polymeric Nanoparticles and Microparticles, CRC
Press, Boca Raton, FL, 1986, pp. 27–93.
[112] T. Daemen, G. Hofstede, M.T. Ten Kate, I.A. Bakker-
[113]
[114]
[115]
[116]
[117]
[118]
[119]
[120]
[121]
[122]
[123]
[124]
Woudenberg, G.L. Scherphof, Liposomal doxorubicin-induced toxicity: depletion and impairment of phagocytic
activity of liver macrophages, Int. J. Cancer 61 (1995)
716–721.
S. Gibaud, C. Rousseau, C. Weingarten, R. Favier, L.
Douay, J.P. Andreux, P. Couvreur, Polyalkylcyanoacrylate
nanoparticles as carriers for granulocyte-colony stimulating
factor (G-CSF), J. Control. Release 52 (1998) 131–139.
N. Kartner, D. Evernden-Porelle, G. Bradley, V. Ling,
Detection of P-glycoprotein in multidrug-resistant cell lines
by monoclonal antibodies, Nature 316 (1985) 820–823.
L. Treupel, M.F. Poupon, P. Couvreur, F. Puisieux, Vectorisation of doxorubicin in nanospheres and reversion of
pleiotropic resistance of tumor cells, C.R. Acad. Sci. III 313
(1991) 171–174.
C. Cuvier, L. Roblot-Treupel, J.M. Millot, G. Lizard, S.
Chevillard, M. Manfait, P. Couvreur, M.F. Poupon, Doxorubicin-loaded nanospheres bypass tumor cell multidrug
resistance, Biochem. Pharmacol. 44 (1992) 509–517.
S. Bennis, C. Chapey, P. Couvreur, J. Robert, Enhanced
cytotoxicity of doxorubicin encapsulated in polyisohexylcyanoacrylate nanospheres against multidrug-resistant
tumour cells in culture, Eur. J. Cancer 1 (1994) 89–93.
`
F. Nemati, C. Dubernet, A. Colin de Verdiere, M.F. Poupon,
L. Treupel Acar, F. Puisieux, P. Couvreur, Some parameters
influencing cytotoxicity of free doxorubicin loaded
nanoparticles in sensitive and multidrug resistant leucemic
murine cells: incubation time, number of particles per cell,
Int. J. Pharm. 102 (1994) 55–62.
A. Colin de Verdiere, C. Dubernet, F. Nemati, M.F. Poupon,
F. Puisieux, P. Couvreur, Uptake of doxorubicin from
loaded nanoparticles in multidrug-resistant leukemic murine
cells, Cancer Chemother. Pharmacol. 33 (1994) 504–508.
A. Colin de Verdiere, C. Dubernet, F. Nemati, E. Soma, M.
Appel, J. Ferte, S. Bernard, F. Puisieux, P. Couvreur,
Reversion
of
multidrug
resistance
with
polyalkylcyanoacrylate nanoparticles: towards a mechanism of
action, Br. J. Cancer 76 (1997) 198–205.
X. Pepin, L. Attali, C. Domrault, S. Gallet, J.M. Metreau, Y.
Reault, P.J. Cardot, M. Imalalen, C. Dubernet, E. Soma, P.
Couvreur, On the use of ion-pair chromatography to
elucidate doxorubicin release mechanism from polyalkylcyanoacrylate nanoparticles at the cellular level, J.
Chromatogr. B 702 (1997) 181–191.
C.E. Soma, C. Dubernet, G. Barratt, F. Nemati, M. Appel,
S. Benita, P. Couvreur, Ability of doxorubicin-loaded
nanoparticles to overcome multidrug resistance of tumor
cells after their capture by macrophages, Pharm. Res. 16
(1999) 1710–1716.
C.E. Soma, C. Dubernet, G. Barratt, S. Benita, P. Couvreur,
Investigation of the role of macrophages on the cytotoxicity
of doxorubicin and doxorubicin-loaded nanoparticles on
M5076 cells in vitro, J. Control. Release 68 (2000) 283–
289.
A. Zimmer, Antisense oligonucleotide delivery with polyhexylcyanoacrylate nanoparticles as carriers, Methods: A
Companion to Methods in Enzymology 18 (1999) 286–
295.
C. Vauthier et al. / Advanced Drug Delivery Reviews 55 (2003) 519–548
[125] C. Chavany, T. Saison-Behmoaras, T. Le Doan, F.
Puisieux, P. Couvreur, C. Helene, Adsorption of oligonucleotides onto polyisohexylcyanoacrylate nanoparticles
protects them against nucleases and increases their cellular
uptake, Pharm. Res. 11 (1994) 1370–1378.
[126] Y. Nakada, E. Fattal, M. Foulquier, P. Couvreur, Pharmacokinetics and biodistribution of oligonucleotide adsorbed onto poly(isobutylcyanoacrylate) nanoparticles after
intravenous administration in mice, Pharm. Res. 13 (1996)
38–43.
[127] G. Schwab, C. Chavany, I. Duroux, G. Goubin, J. Lebeau,
C. Helene, T. Saison-Behmoaras, Antisense oligonucleotides adsorbed to polyalkylcyanoacrylate nanoparticles specifically inhibit mutated Ha-ras-mediated cell proliferation
and tumorigenicity in nude mice, Proc. Natl. Acad. Sci.
USA 91 (1994) 10460–10464.
[128] G. Lambert, E. Fattal, A. Brehier, J. Feger, P. Couvreur,
Effect of polyisobutylcyanoacrylate nanoparticles and
lipofectin loaded with oligonucleotides on cell viability and
PKC alpha neosynthesis in HepG2 cells, Biochimie 80
(1998) 969–976.
[129] G. Lambert, E. Fattal, H. Pinto-Alphandary, A. Gulik, P.
Couvreur, Polyisobutylcyanoacrylate nanocapsules containing an aqueous core for the delivery of oligonucleotides,
Int. J. Pharm. 214 (2001) 13–16.
[130] G. Lambert, J.R. Bertrand, E. Fattal, F. Subra, H. PintoAlphandary, C. Malvy, C. Auclair, P. Couvreur, EWS fli-1
antisense nanocapsules inhibit Ewing sarcoma-related
tumor in mice, Biochem. Biophys. Res. Commun. 279
(2000) 401–406.
[131] K. Tanaka, T. Iwakuma, K. Harimaya, H. Sato, Y. Iwamoto,
EWS-Fli1 antisense oligodeoxynucleotide inhibits proliferation of human Ewing’s sarcoma and primitive neuroectodermal tumor cells, J. Clin. Invest. 99 (1997) 239–247.
[132] C.A. Stein, Phosphorothioate antisense oligodeoxynucleotides: questions of specificity, Biotechnology 14 (1996)
47–149.
[133] J. Kreuter, Nanoparticulate systems for brain delivery of
drugs, Adv. Drug Deliv. Rev. 47 (2001) 65–81.
[134] J. Kreuter, R.N. Alyautdin, D.A. Kharkevich, A.A. Ivanov,
Passage of peptides through the blood–brain barrier with
colloidal polymer particles (nanoparticles), Brain Res. 674
(1995) 171–174.
[135] R.N. Alyautdin, V.E. Petrov, K. Langer, A. Berthold, D.A.
Kharkevich, J. Kreuter, Delivery of loperamide across the
blood–brain barrier with polysorbate 80-coated polybutylcyanoacrylate nanoparticles, Pharm. Res. 14 (1997)
325–328.
[136] R.N. Alyautdin, E.B. Tezikov, P. Ramge, D.A. Kharkevich,
D.J. Begley, J. Kreuter, Significant entry of tubocurarine
into the brain of rats by adsorption to polysorbate 80-coated
polybutylcyanoacrylate nanoparticles: an in situ brain perfusion study, J. Microencapsul. 15 (1998) 67–74.
[137] A. Friese, E. Seiller, G. Quack, B. Lorenz, J. Kreuter,
Increase of the duration of the anticonvulsive activity of a
novel NMDA receptor antagonist using poly(butylcyanoacrylate) nanoparticles as a parenteral controlled
[138]
[139]
[140]
[141]
[142]
[143]
[144]
[145]
[146]
[147]
[148]
[149]
[150]
547
release system, Eur. J. Pharm. Biopharm. 49 (2000) 103–
109.
M. Sopala, S. Schweizer, N. Schafer, E. Nurnberg, J.
Kreuter, E. Seiller, W. Danysz, Neuroprotective activity of a
nanoparticulate formulation of the glycine B site antagonist
MRZ 2 / 576 in transient focal ischaemia in rats, Arzneimittelforschung 52 (2002) 168–174.
A.E. Gulyaev, S.E. Gelperina, I.N. Skidan, A.S. Antropov,
G.Y. Kivman, J. Kreuter, Significant transport of doxorubicin into the brain with polysorbate 80-coated nanoparticles,
Pharm. Res. 16 (1999) 1564–1569.
S.E. Gelperina, A.S. Khalansky, I.N. Skidan, Z.S. Smirnova, A.I. Bobruskin, S.E. Severin, B. Turowski, F.E.
Zanella, J. Kreuter, Toxicological studies of doxorubicin
bound to polysorbate 80-coated poly(butyl cyanoacrylate)
nanoparticles in healthy rats and rats with intracranial
glioblastoma, Toxicol. Lett. 126 (2002) 131–141.
P. Ramge, R.E. Unger, J.B. Oltrogge, D. Zenker, D. Begley,
J. Kreuter, H. Von Briesen, Polysorbate-80 coating enhances
uptake of polybutylcyanoacrylate (PBCA)-nanoparticles by
human and bovine primary brain capillary endothelial cells,
Eur. J. Neurosci. 12 (2000) 1931–1940.
R.N. Alyaudtin, A. Reichel, R. Lobenberg, P. Ramge, J.
Kreuter,
D.J.
Begley,
Interaction
of
poly(butylcyanoacrylate) nanoparticles with the blood–brain
barrier in vivo and in vitro, J. Drug Target. 9 (2001)
209–221.
L. Araujo, R. Lobenberg, J. Kreuter, Influence of the
surfactant concentration on the body distribution of
nanoparticles, J. Drug Target. 6 (1999) 373–385.
J.C. Olivier, L. Fenart, R. Chauvet, C. Pariat, R. Cecchelli,
W. Couet, Indirect evidence that drug brain targeting using
polysorbate 80-coated polybutylcyanoacrylate nanoparticles
is related to toxicity, Pharm. Res. 16 (1999) 1836–1842.
P. Calvo, B. Gouritin, H. Villarroya, F. Eclancher, C.
Giannavola, C. Klein, J.P. Andreux, P. Couvreur, Quantification and localization of PEGylated polycyanoacrylate
nanoparticles in brain and spinal cord during experimental
allergic encephalomyelitis in the rat, Eur. J. Neurosci. 15
(2002) 1317–1326.
P. Jani, G.W. Halbert, J. Langridge, A.T. Florence, The
uptake and translocation of latex nanospheres and microspheres after oral administration to rats, J. Pharm. Pharmacol. 41 (1989) 809–812.
H.E. Le Fevre, D.D. Joel, G. Shidlousky, Retention of
ingested latex particles in Peyers patches of gerinfree and
conventional mice, Soc. Exp. Biol. Med. 179 (1985) 522–
528.
C. Damge, C. Michel, M. Aprahamian, P. Couvreur, J.P.
Devissaguet, Nanocapsules as carriers for oral peptide
delivery, J. Control. Release 13 (1990) 233–239.
A.T. Florence, N. Hussain, Transcytosis of nanoparticle and
dendrimer delivery systems: evolving vistas, Adv. Drug
Deliv. Rev. 50 (2001) S69–S89.
J. Kreuter, V. Muller, K. Munz, Quantitative and microautoradiographic study on mouse intestinal distribution
of polycyanoacrylate nanoparticles, Int. J. Pharm. 55 (1989)
39–45.
548
C. Vauthier et al. / Advanced Drug Delivery Reviews 55 (2003) 519–548
[151] M. Aprahamian, C. Michel, W. Humbert, J.P. Devissaguet,
C. Damge, Transmucosal passage of polyalkylcyanoacrylate
nanocapsules as a new drug carrier in the small intestine,
Biol. Cell 61 (1987) 69–76.
[152] M. Aboubakar, P. Couvreur, H. Pinto-Alphandary, B.
Gouritin, B. Lacour, R. Farinotti, F. Puisieux, C. Vauthier,
Insulin-loaded nanocapsules for oral administration: in vitro
and in vivo investigation, Drug Deliv. Res. 49 (2000)
109–117.
[153] C. Damge, H. Vranckx, P. Balschmidt, P. Couvreur, Poly(alkyl cyanoacrylate) nanospheres for oral administration of
insulin, J. Pharm. Sci. 86 (1997) 1403–1409.
[154] S. Watnasirichaikul, T. Rades, I.G. Tucker, N.M. Davies,
In-vitro release and oral bioavailability in diabetic rats of
insulin using nanocapsules dispersed in biocompatible
microemulsion, J. Pharm. Pharmacol. 54 (2002) 473–480.
[155] C. Damge, J. Vonderscher, P. Marbach, M. Pinget, Poly(alkyl cyanoacrylate) nanocapsules as a delivery system in
the rat for octreotide, a long-acting somatostatin analogue,
J. Pharm. Pharmacol. 49 (1997) 949–954.
[156] M. Torres-Lugo, N.A. Peppas, Transmucosal delivery systems for calcitonin: a review, Biomaterials 21 (2000) 1191–
1196.
[157] D.T. O’Hagan, K. Palin, S.S. Davis, Poly(butyl-2cyanoacrylate) particles as adjuvant for oral immunization,
Vaccine 7 (1989) 213–216.
[158] G. Ponchel, F. Touchard, D. Duchene, N.A. Peppas,
Bioadhesive analysis of the controlled-release systems. III.
Fracture and interpenetration analysis in poly(acrylic acid)
containing systems, J. Control. Release 5 (1987) 129–141.
[159] G. Ponchel, J. Irache, Specific and non-specific bioadhesive
particulate systems for oral delivery to the gastrointestinal
tract, Adv. Drug Deliv. Rev. 34 (1998) 191–219.
[160] H. Takeuchi, H. Yamamoto, Y.Y. Kawashima, Mucoadhesive nanoparticulate systems for peptide drug delivery, Adv.
Drug Deliv. Rev. 47 (2001) 39–54.
[161] C. Pimienta, F. Chouinard, A. Labib, V. Lenaerts, Effect of
various poloxamer coatings on in vitro adhesion of isohexylcyanoacrylate nanospheres to rat ileal segments under
liquid flow, Int. J. Pharm. 80 (1992) 1–8.
[162] V. Lenaerts, P. Couvreur, L. Grislain, P. Maincent,
Nanoparticles as a gastrointestinal drug delivery system, in:
V. Lenaerts, R. Gurny (Eds.), Bioadhesive Drug Delivery
Systems, CRC Press, Boca Raton, FL, 1990, pp. 94–104.
[163] P. Maincent, R. Le Verge, P. Sado, P. Couvreur, J.P.
Devissaguet, Disposition kinetics and oral bioavailability of
vincamine-loaded polyalkyl cyanoacrylate nanoparticles, J.
Pharm. Sci. 75 (1986) 955–958.
[164] C. Damge, M. Aprahamian, G. Balboni, A. Hoeltzel, V.
Andrieu, J.P. Devissaguet, Polyalkylcyanoacrylate nanocapsules increase the intestinal absorption of a lipophilic drug,
Int. J. Pharm. 36 (1987) 121–125.
[165] B. Hubert, J. Atkinson, M. Guerret, M. Hoffman, J.P.
Devissaguet, P. Maincent, The preparation and acute antihypertensive effects of a nanocapsular form of darodipine, a
dihydropyridine calcium entry blocker, Pharm. Res. 8
(1991) 734–738.
[166] H. Boudad, P. Legrand, M.H. Coconnier, M. Appel, D.
Duchene, G. Ponchel, Absorption of saquinavir from combined hydroxypropyl b cyclodextrin and poly(alkylcyanoacrylate) nanoparticles through CACO-2 cells
monolayers, in: Proceedings of the 4th World Meeting on
Pharmaceutics Biopharmaceutics Pharmaceutical Technology, Florence, Italy, 8–11 April, 2001, pp. 1385–1386.
[167] J.C. Gautier, J.L. Grangier, A. Barbier, P. Dupont, D.
Dussosoy, G. Pastor, P. Couvreur, Biodegradable nanoparticles for subcutaneous administration of growth hormone
releasing factor (hGRF), J. Control. Release 3 (1992) 205–
210.
[168] A. Zimmer, J. Kreuter, J.R. Robinson, Studies on the
transport pathway of PBCA nanoparticles in ocular tissues,
J. Microencapsul. 8 (1991) 497–504.
[169] C.A. Le Bourlais, F. Chevanne, B. Turlin, L. Acar, H. Zia,
P.A. Sado, T.E. Needham, R. Leverge, Effect of cyclosporine A formulations on bovine corneal absorption: ex-vivo
study, J. Microencapsul. 14 (1997) 457–467.
[170] S.D. Desai, J. Blanchard, Pluronic F127-based ocular
delivery systems containing biodegradable polyisobutylcyanoacrylate nanocapsules of pilocarpine, J. Deliv.
Target. Ther. Agents 7 (2000) 201–207.
[171] M. Fresta, G. Fontana, C. Bucolo, G. Cavallaro, G.
Giammona, G. Puglisi, Ocular tolerability and in vivo
bioavailability of poly(ethylene glycol) (PEG)-coated
polyethyl-2-cyanoacrylate
nanospheres-encapsulated
acyclovir, J. Pharm. Sci. 90 (2001) 288–297.