Sunteți pe pagina 1din 18

Pharmacological Reports Copyright 2012

2012, 64, 10201037 by Institute of Pharmacology


ISSN 1734-1140 Polish Academy of Sciences

Review

Nanoparticles as drug delivery systems


Agnieszka Z. Wilczewska1, Katarzyna Niemirowicz2, Karolina H. Markiewicz1,
Halina Car2

Institute of Chemistry, University of Bialystok, al. J. Pilsudskiego 11/4, PL 15-433 Biaystok, Poland

Department of Experimental Pharmacology, Medical University of Bialystok, Szpitalna 37, PL 15-295 Biaystok,
Poland

Correspondence: Halina Car, e-mail: hcar@umb.edu.pl (concerning medical applications);


Agnieszka Z. Wilczewska, e-mail: agawilcz@uwb.edu.pl (concerning chemistry of nanoparticles)

Abstract:
Controlled drug delivery systems (DDS) have several advantages compared to the traditional forms of drugs. A drug is transported to
the place of action, hence, its influence on vital tissues and undesirable side effects can be minimized. Accumulation of therapeutic
compounds in the target site increases and, consequently, the required doses of drugs are lower. This modern form of therapy is espe-
cially important when there is a discrepancy between the dose or the concentration of a drug and its therapeutic results or toxic ef-
fects. Cell-specific targeting can be accomplished by attaching drugs to specially designed carriers. Various nanostructures,
including liposomes, polymers, dendrimers, silicon or carbon materials, and magnetic nanoparticles, have been tested as carriers in
drug delivery systems. In this review, the aforementioned nanocarriers and their connections with drugs are analyzed. Special atten-
tion is paid to the functionalization of magnetic nanoparticles as carriers in DDS. Then, the advantages and disadvantages of using
magnetic nanoparticles as DDS are discussed.

Key words:
drug delivery system, nanocarriers, nanoparticles, magnetic nanoparticles, targeting therapy

Introduction of drug are required [111]. This modern form of ther-


apy is especially important when there is a discrep-
Delivering therapeutic compound to the target site is ancy between a dose or concentration of a drug and its
a major problem in treatment of many diseases. therapeutic results or toxic effects.
A conventional application of drugs is characterized Cell-specific targeting can be achieved by attach-
by limited effectiveness, poor biodistribution, and ing drugs to individually designed carriers. Recent de-
lack of selectivity [111]. These limitations and draw- velopments in nanotechnology have shown that nano-
backs can be overcome by controlling drug delivery. particles (structures smaller than 100 nm in at least
In controlled drug delivery systems (DDS) the drug is one dimension) have a great potential as drug carriers.
transported to the place of action, thus, its influence Due to their small sizes, the nanostructures exhibit
on vital tissues and undesirable side effects can be unique physicochemical and biological properties
minimized. In addition, DDS protects the drug from (e.g., an enhanced reactive area as well as an ability to
rapid degradation or clearance and enhances drug cross cell and tissue barriers) that make them a favor-
concentration in target tissues, therefore, lower doses able material for biomedical applications.

1020 Pharmacological Reports, 2012, 64, 10201037


Nanoparticles and drugs
Agnieszka Z. Wilczewska et al.

The way of conjugating the drug to the nanocarrier


Nanocarriers used in drug delivery system
and the strategy of its targeting is highly important for
a targeted therapy. A drug may be adsorbed or cova-
According to the definition from NNI (National lently attached to the nanocarriers surface or else it
Nanotechnology Initiative), nanoparticles are struc- can be encapsulated into it. Covalent linking has the
tures of sizes ranging from 1 to 100 nm in at least one advantage over other ways of attaching as it enables
dimension. However, the prefix nano is commonly to control the number of drug molecules connected to
used for particles that are up to several hundred the nanocarrier, i.e., a precise control of the amount of
nanometers in size. therapeutic compound delivered. Cell-specific target-
Nanocarriers with optimized physicochemical and ing with nanocarriers may be accomplished by using
biological properties are taken up by cells more easily active or passive mechanisms. The first strategy relies
than larger molecules, so they can be successfully on the attraction of a drug the nanocarriers conju-
used as delivery tools for currently available bioactive gate to the affected site by using recognition ligands,
compounds [145]. Liposomes, solid lipids nanoparti- attached to the surface of conjugates antibodies, low
cles, dendrimers, polymers, silicon or carbon materi- molecular ligands, e.g., folic acids, peptides, etc. The
als, and magnetic nanoparticles are the examples of active strategy can be also achieved through a ma-
nanocarriers that have been tested as drug delivery nipulation of physical stimuli (e.g., temperature, pH,
systems (Fig. 1). magnetism). Passive targeting is a result of enhanced

Fig. 1. Nanoparticle drug delivery systems with relation to other scales

Pharmacological Reports, 2012, 64, 10201037 1021


vascular permeability and retention (EPR) which is cancer activity [48]. A drug is incorporated in lipo-
characteristic of leaky tissues of tumors [111]. somes by the encapsulation process (Fig. 2). The
Once the drug-nanocarrier conjugates reach the release of a drug from liposomes depends on the lipo-
diseased tissues, the therapeutic agents are released. some composition, pH, osmotic gradient, and the sur-
A controlled release of drugs from nanocarriers can rounding environment [48]. Additionally, a prolonged
be achieved through changes in physiological envi- residence time increases the duration of action of such
ronment such as temperature, pH, osmolality, or via particles, but decreases their number. Interactions of
an enzymatic activity. liposomes with cells can be realized by: adsorption,
Nanocarriers used for medical applications have to fusion, endocytosis, and lipid transfer. There are a lot
be biocompatible (able to integrate with a biological of drug examples in liposomal formulations, such as
system without eliciting immune response or any anticancer drugs [48], neurotransmitters (serotonin)
negative effects) and nontoxic (harmless to a given [2], antibiotics [155, 175], anti-inflammatory [113],
biological system). Undesirable effects of nanoparti- and antirheumatic drugs [158]. Recent studies have
cles strongly depend on their hydrodynamic size, reported the clinical outcomes and side effects of pho-
shape, amount, surface chemistry, the route of admini- todynamic therapy (PDT) by means of intense pulsed
stration, reaction of the immune system (especially light (IPL) and spray (liposome encapsulated 0.5%
a route of the uptake by macrophages and granulo- 5-aminolevulinic acid) which was used for the treat-
cytes) and residence time in the bloodstream. Due to ment of inflammatory facial acne [171]. Turkova et al.
a number of factors which may affect the toxicity of [155] compared the efficacy and safety of deoxycho-
nanoparticles, their estimation is rather difficult and, late and lipid (liposomal) amphotericin B formula-
thus, toxicological studies of each new DDS formula- tions (AMBF) in the treatment of invasive fungal dis-
tion are needed. However, with respect to their size, ease (IFD) in neonates. The authors of the study have
one can make some generalizations smaller particles reported that deoxycholate amphotericin B is cheap
have a greater surface area, thus, they are more reac- and effective in treating neonatal IFD. The therapy
tive and, in consequence, more toxic [5]. It is gener- appears to be safe for use as a first-line therapy if the
ally accepted that nanoparticles with a hydrodynamic underlying risk for nephrotoxicity is low [155]. Saf-
diameter of 10100 nm have optimal pharmacokinetic dar and co-workers [130] conducted a meta-analysis
properties for in vivo applications. Smaller nanoparti-
in order to evaluate nephrotoxicity associated with
cles are subjects to tissue extravasations and renal
amphotericin B lipid complex (ABLC) and liposomal
clearance whereas larger are quickly opsonized and
amphotericin B (L-AmB). They showed that nephro-
removed from the bloodstream via the macrophages
toxicity is generally similar for ABLC and L-AmB in
of the reticuloendothelial system [35].
patients receiving antifungal therapy and prophylaxis
[130]. The unresolved problem of using drug delivery
systems based on liposomes arises from their accumu-
lation in cells (liver macrophages) outside the target
Liposomes
tissues and the unpredictable effects dependent on the
active agent they carry, such as cellular death [41].
Liposomes have been the first to be investigated as Modified liposomes are an interesting type of such
drug carriers. They are nano/micro-particular or col- lipid structures. The multifunctional liposomes, con-
loidal carriers, usually with 80300 nm size range taining the specific proteins, antigens, or other bio-
[144]. They are spherical vesicles composed of phos- logical substances, can be used to design drugs which
pholipids and steroids (e.g., cholesterol), bilayers, or act selectively on a particular tissue. It is a promising
other surfactants and form spontaneously when cer- approach for targeted delivery of therapeutics. Biswas
tain lipids are dispersed in aqueous media where lipo- et al. [22] presented hydrazine-functionalized poly-
somes can be prepared, e.g., by sonication [139]. (ethylene glycol)-phosphatidylethanolamine (PEG-PE)-
Liposomes have been reported to increase the solubil- based amphiphilic polymer which can conjugate a va-
ity of drugs and improve their pharmacokinetic prop- riety of ligands. The researchers investigated the re-
erties, such as the therapeutic index of chemothera- versible model ligands monoclonal antinucleosome
peutic agents, rapid metabolism, reduction of harmful antibody 2C5 and antimyosin antibody 2G4, as well
side effects and increase of in vitro and in vivo anti- as glycoproteins concanavalin A (Con-A). The re-

1022 Pharmacological Reports, 2012, 64, 10201037


Nanoparticles and drugs
Agnieszka Z. Wilczewska et al.

versible attachment of homing devices is useful espe- i.e., lipids solid at the body temperature [165]. They
cially in modified liposomal systems, whereafter they have been exploited for the dermal [1], peroral [100],
successfully perform the function of targeting at the parenteral [109], ocular [13], plumonary [83], and
specific site. Ligands, such as antibodies, are cleaved rectal [147] delivery.
off in response to an environmental stimulus, e.g., pH SLN are particles made of solid lipids, e.g., highly
[22]. In addition, cationic liposomes (CLs) can be purified triglycerides, complex glyceride mixtures or
used as a gene delivery carrier [167]. They are better waxes stabilized by various surfactants [76]. The
than natural or anionic liposomes for gene transfer main characteristics of SLN include a good physical
[167]. Kim and co-workers [72] studied modified stability, protection of incorporated drugs from degra-
cationic liposomes either by polyethylene glycol dation, controlled drug release, and good tolerability.
(PEG)-grafting or PEG-adding methods as transfec- Additionally, some disadvantages have been ob-
tion complexes of plasmid DNA. In a recent study, served, such as low loading capacity (limited by the
Biswas et al. [21] have examined polyethylene solubility of drug in the lipid and the structure and
glycol-phosphatidylethanolamine (PEG-PE) conju- polymorphic state of the lipid matrix), drug expulsion
gate with the TPP group as drug carriers. They used after crystallization, and a relatively high water con-
paclitaxel (PTX) as a model drug and studied them for tent of the dispersions [104, 165].
their toxicity, mitochondrial targeting, and efficacy in NLC and LDC are modifications of lipid based
delivering. As a result, they suggested that TPP- nanoparticles that have been developed to overcome
PEG-PE can be used as non-toxic, mitochondria- limitations of conventional SLN. NLC are produced
targeted drug delivery systems [21]. by mixing solid lipids with liquid lipids, which leads
to special nanostructure with increased payload and
prevented drug expulsion. Three types of NLC have
been introduced: imperfect type NLC (general imper-
fections in the matrix nanostructure form free spaces
Nanoparticles based on solid lipids
for the accommodation of the guest molecules), mul-
tiple type NLC (drugs are solved in oils and protected
SLN (solid lipid nanoparticles), NLC (nanostructured from degradation by the surrounding solid lipid) and
lipid carriers) and LDC (lipid drug conjugates) are amorphous type NLC (the crystallization that causes
types of carrier systems based on solid lipid matrix, drug expulsion is avoided) [157]. NLC are mainly ex-

Fig. 2. Drug delivery by liposomes

Pharmacological Reports, 2012, 64, 10201037 1023


ploited for dermal applications [103, 125]. LDC were PNPs are usually coated with nonionic surfactants
developed in order to expand applicability of lipid in order to reduce immunological interactions (e.g.,
based carriers to lipophobic drug molecules. These in- opsonization or presentation PNPs to CD8 T-lympho-
soluble drug-lipid conjugates can be prepared by salt cytes) as well as intermolecular interactions between
formation or by covalent linking followed by homog- the surface chemical groups of PNPs (e.g., van der
enization [102, 165]. Waals forces, hydrophobic interaction or hydrogen
bonding) [152].
Drugs can be immobilized on PNPs surface after
a polymerization reaction [87] or can be encapsulated
on PNP structure during a polymerization step [99].
Polymeric nanoparticles
Moreover, drugs may be released by desorption,
diffusion, or nanoparticle erosion in target tissue
Polymeric nanoparticles (PNPs) are structures with [152]. The examples of drug-polymeric nanocarrier
a diameter ranging from 10 to 100 nm. The PNPs are conjugates used as drug delivery systems are shown
obtained from synthetic polymers, such as poly- in Table 1.
e-caprolactone [20], polyacrylamide [14] and poly- Among the aforementioned applications the one
acrylate [156], or natural polymers, e.g., albumin that is particularly interesting is the immobilization of
[94], DNA [93], chitosan [93, 128] gelatin [131]. retinyl acetate (RA) on ethyl cellulose (EC), which
Based on in vivo behavior, PNPs may be classified as improves aqueous stability and photostability of
biodegradable, i.e., poly(L-lactide) (PLA) [91], poly- a drug. In ex vivo tests on a skin tissue of mice,
glycolide (PGA) [118], and non-biodegradable, e.g., a 100% absorption of RA after 24 h has been demon-
polyurethane [55]. strated [8]. It is also worth to point out the biodegrad-

Tab. 1. Polymer nanocarriers as DDS

Drug Therapeutic activity Nanocarrier Ref.

Carboplatin Antineoplastic drug, ovarian, head, neck and lung cancer Sodium alginate 106
5-Fluorouracil (5-FU) Anticancer drug, colon cancer CS-g-poly(N-vinyl 128
caprolactam)
Doxorubicin Antineoplastic agent, wide spectrum of tumors PEGylatedPLGA 118
Capecitabine Pro-drug of fluorouracil, metastatic colorectal and breast CS-poly(ethylene 3
cancer oxide-g-acrylamide)

Mitomycin C Chemotherapeutic agent, bladder tumors CS, CS-PCL, PLL-PCL 20

Rifampicin Antitubercular drugs, latent M. tuberculosis infection Gelatin 131


in adults
Cyclosporine A Cyclic polypeptide, immunosuppressant GMO/poloxamer 407 cubic 79
nanoparticles
Lamivudine Anti-HIV drug PLA/CS 43
Tacrine Anti-Alzheimer drug CS 164
Retinyl acetate Photoaging, severe acne and skin inflammation EC 8
Analogue of b-lactam Antibiotic, infection G(+)bacteria Polyacrylate 156
Clotrimazole Antifungal drug PLA-co-PLG 115

CS chitosan; PEGylatedPLGA PEGylated poly(lactic-co-glycolic acid); PCL polycaprolactone; PLL poly(L-lysine); GMO glyceryl
monooleate; EC ethyl cellulose, PLA polylactide; PLG polyglycolide

1024 Pharmacological Reports, 2012, 64, 10201037


Nanoparticles and drugs
Agnieszka Z. Wilczewska et al.

able thermo-responsive chitosan-g-poly(N-vinylcapro- drimers and their so-called polyvalence is particularly


lactam)-biopolymer used for the delivery of 5-fluoro- relevant for biomedical purposes. Dendrimers cyto-
uracil to cancer cells. The hypothesized mechanism of toxicity depends on the core material and is strongly
5-FU controlled release from this polymeric nanocar- influenced by the nature of the dendrimers surface.
rier is swelling followed by conformational changes For example, changing the surface amine groups into
during a LCST (lower critical solution temperature) hydroxyl ones may result in lower levels of cytotoxic-
transition. The in vitro drug release showed a signifi- ity. The term polyvalence defines the number of active
cant release above LCST. The high toxicity to cancer groups on a dendrimers surface. The presence of sev-
cells, comparatively lower to the normal ones, was eral surface functional groups enables a sumultanoeus
observed [128]. interaction with a number of receptors, thus, it en-
The application of biodegradable nanosystems for hances biological activity.
the development of nanomedicines is one of the most There are a few ways of connecting biologically
successful ideas. Nanocarriers composed of biode- active compounds to dendrimers. The drug may be
gradable polymers undergo hydrolysis in the body, encapsulated in the internal structure of dendrimers
producing biodegradable metabolite monomers, such [40] or it can be chemically attached or physically ad-
as lactic acid and glycolic acid. Kumari et al. [78] re- sorbed on dendrimers surface [97]. The choice of the
ported a minimal systemic toxicity associated with us- immobilization method depends on the drug proper-
ing of PLGA for drug delivery or biomaterial applica- ties. Encapsulation is used when drugs are labile,
tions. Such nanoparticles are biocompatible with tis- toxic, or poorly soluble. In turn, chemical attachment
sue and cells [116]. Drug-biodegradable polymeric provides the possibility to control quantity of drugs on
nanocarrier conjugates used for drug delivery are sta- dendrimers surface by controlling the number of co-
ble in blood, non-toxic, and non-thrombogenic. They valent bonds [140]. The selectivity of both methods
are also non-immunogenic as well as non-proinflam- may be enhanced by attaching on the dendrimers sur-
matory, and they neither activate neutrophils nor af- face such targeting agents as folic acid [40, 140] or
fect reticuloendothelial system [42]. epidermal growth factor [173].
The surface of dendrimers provides an excellent
platform for an attachment of specific ligands, which
may include folic acid [140], antibodies [161], cyclic
Dendrimer nanocarriers targeting peptides arginine-glycine-aspartic acid
(RGD) [159], selective A3 adenosine receptor [153],
Dendrimers are unique polymers with well-defined silver salts complexes antimicrobial agents [16], or
size and structure. Dendritic architecture is one of the poly(ethylene glycol) (PEG) [85]. The attached com-
most popular structures observed throughout all bio- pounds can improve surface activity as well as the
logical systems. Some of the examples of nanometric biological and physical properties of dendrimers.
molecules possessing dendritic structure include: gly- Poly(amido amide) (PAMAM) is a dendrimer
cogen, amylopectin, and proteoglycans [146]. which is frequently used in biomedical applications.
In the structure of dendrimer, in contrast to the lin- Both the structure of PAMAM dendrimers and the
ear polymer, the following elements can be distin- distribution of drugs [135] or genes [173] inside these
guished: a core, dendrons, and surface active groups. molecules have been intensively investigated. PAMAM
The core is a single atom or molecule (only if it has at dendrimers grow through generations G = 110 and
least two identical functional groups) that dendrons their size increases from 1.1 to 12.4 nm. The size of
are attached to. The dendrons (dendrimer arms) are the respective generations of PAMAM is comparable
molecules of monomer linked with the core, forming to the size of selected proteins, e.g., dendrimer G3
layers and building successive generations (their with insulin and G7 with hemerythrin [150]. An ex-
growth is spatially limited). Biocompatibility and ample of a drug immobilization in PAMAM dendri-
physicochemical properties of dendrimers are deter- mer is cisplatin. This complex compared with free
mined by surface functional groups [23]. cisplatin exhibits several advantages, such as slower
Selection of a core, type of a monomer and surface rate of drug release, higher accumulation of the drug
functional groups determine the usability of den- in solid tumors, and lower toxicity in all organs
drimers in medical applications. Cytotoxicity of den- [40, 92]. Further examples of drug incorporation in

Pharmacological Reports, 2012, 64, 10201037 1025


PAMAM dendrimers are anticancer drugs, including blood circulation and an avoidance of dendrimers ac-
methotrexate [108], doxorubicin [59], 5-FU [140], cumulation in normal organs, such as kidneys and
and anti-inflammatory drugs, e.g., ibuprofen [149], liver [75].
piroxicam [122], or indomethacin [26].
The size and charge of PAMAM dendrimers influ-
ence their cytotoxicity. The higher-generation (G4-
G8) PAMAM dendrimers exhibit toxicity due to their Silica materials
high cationic charge density [134]. Comparative tox-
icity studies on cationic and anionic dendrimers using
Caco-2 cells have shown a significantly lower cyto- Silica materials used in controlled drug delivery sys-
toxicity of anionic compounds in comparison with the tems are classified as xerogels [36] and mesoporous
cationic ones [74]. Positively charged dendrimers in- silica nanoparticles (MSNs), e.g., MCM-41 (Mobil
troduced into the systemic circulation interact with Composition of Matter) and SBA-15 (Santa Barbara
blood components causing destabilization of cell University mesoporous silica material) [163]. They ex-
membranes and cell lysis [63]. Roberts and co- hibit several advantages as carrier systems, including
workers [129] observed that cationic PAMAM den- biocompatibility, highly porous framework and an ease
drimers caused a decrease in cell viability, however, in terms of functionalization [7]. Among inorganic
they found no evidence of their immunogenicity in nanoparticles, silica materials are the carriers which
rabbits. Additionally, they studied the toxicity of cati- most often are chosen for biological purposes [141].
onic PAMAM Starburst in mice and they suggested Silica xerogels possess an amorphous structure with
high porosity and enormous surface area. A porous
that even high doses of low generation cationic den-
structure (shape and pore dimensions) depends on syn-
drimers do not cause side effects. Recent studies have
thesis parameters [50]. Sol-gel technique is frequently
shown that G4 dendrimers, which have amino termi-
used to form silica xerogels loaded with drugs.
nal groups, are toxic and impair the growth and devel-
A modification of the synthesis conditions, such as the
opment of zebrafish embryos. In turn, dendrimers
ratio of reagents, temperature, concentration of the
with carboxylic acid functional groups did not exert
catalyst, and pressure of drying, allows to alter proper-
adverse effect on zebrafish embryos [61]. Dendrimers
ties of xerogels used in controlled drug release [36,
can modulate cytokine and chemokine release. This
126]. Phenytoin [52], doxorubicin [124], cisplatin [38],
property turned out to be helpful in therapy, but it can
metronidazole [39], nifedipine [95], diclofenac [37],
also cause serious toxic effects [49]. The PAMAM and heparin [4] are examples of drugs which have been
generation of 3,5-glucosamine dendrimers induces loaded into xerogels using this technique.
a synthesis of pro-inflammatory chemokines, such as The best known types of mesoporous silica nano-
MIP-1a, MIP-1h, and cytokines TNF-a, IL-1h, IL-6, materials are MCM-41 with a hexagonal arrangement
IL-8 in human dendritic cells and macrophages, of the mesopores and SBA-15 with a well-ordered
which exhibits an immunomodulatory effect. Chau- hexagonal connected system of pores [163]. The
han et al. [27] studied in vivo toxicity profile of MSNs, in comparison with xerogels, possess more
PAMAM dendrimers in mice. The researchers as- homogenous structure, lower polydispersity and
sessed the following parameters: the animal behavior, higher surface area for adsorption of therapeutic or di-
feed intake, body weight, carbohydrate, lipid and pro- agnostic agents [46]. The mechanism of drug loading
tein metabolism, hematological parameters, histopa- into mesoporous silica material is a chemical or
thology, and cell viability. They observed no effect on physical adsorption [46]. By these processes, diverse
other hematological (excluding red blood cells, hema- types of drugs, including anticancer drugs [46, 60],
tocrit value and hemoglobin) and biochemical pa- antibiotics [81], and heart disease drugs [121], have
rameters (excluding the decrease of glucose levels in been embedded into MNSs. The drug release is usu-
the high-NH dose) as well as on feed intake, body ally controlled by diffusion [81]. The silicalites and
and organ weights. In addition, the histopathology mesoporous silica nanoparticles potential application
showed a toxic effect on liver and kidneys [27]. PE- in photodynamic therapy has been also studied [62].
Gylation of dendritic systems is a way of lowering The MSNs properties make them an excellent ma-
general toxicity. This process enables a long-lasting terial for various pharmaceutical and biomedical ap-

1026 Pharmacological Reports, 2012, 64, 10201037


Nanoparticles and drugs
Agnieszka Z. Wilczewska et al.

plications. The structure of MSNs enables the incor- The size of nanoparticles plays a key role in their
poration of both small [46] and large molecules [73], toxicity. Cho et al. [32] investigated the effect of the
adsorption of DNA, and gene transfer [142]. This particle size on the pharmacokinetic parameters, such
gives a possibility of using these nanomaterials in as tissue distribution and excretion via intravenously
a combined therapy [60]. injected silica particles of different sizes in mice.
Some data indicate that nano-sized silica particles They observed an occurrence of inflammatory re-
(SNPs) are biocompatible and have a great potential sponse of the liver within 12 h after injection of 200
for a variety of diagnostic and therapeutic applica- and 100 nm silica nanoparticles. However, this effect
tions in medicine. However, recent studies have re- was not reported for the smaller particles (50 nm). All
vealed in vitro and in vivo toxicity and certain hazards types of particles were excreted via urine and bile.
of using nanosilica. Most of the in vitro studies of These three types of nanoparticles were accumulated
silica nanoparticles show the adverse effect in investi- by macrophage in liver and spleen and remained there
gated cells. The described effect depends on the cell up to 4 weeks after the first injection of a single dose
type and nanoparticle size. Silica nanoparticles have [32]. The authors then presented the effect of amor-
an impact on a generation of oxidative stress in cells phous silica by intratracheal instillation. They ob-
via formation of reactive oxygen species [84], ele- served significant increase of lung weights, total
vated production of malondialdehyde [82], decreasing number of BAL cells and proteins concentration. The
glutathione level [174], and induction of antioxidant histopathological evaluation showed an inflammation,
enzymes, including superoxide dismutase (SOD) and characterized by late neutrophils infiltration which re-
heme oxygenase 1 (OH-1) [117]. All of these events sulted in the occurrence of symptoms of chronic
are responsible for lipid peroxidation and cell mem- granulomatous inflammation of the lung [33]. Other
brane damage [82]. Previous work showed that expo- researchers reported that inhalation of silica nanopar-
sure to silica nanoparticles at high concentrations ticles caused the inflammation of pulmonary tract
caused activation of NF-kB in endothelial cells [84] myocardial ischemic damage and atrio-ventricular
or Nrf-2-ERK MAP kinase signaling pathway in hu- blockage. In addition, they observed an increase of fi-
man bronchial epithelial cells [51]. Pro-inflammatory brinogen concentration in blood [30]. Although or-
response resulted in an induction of various chemoki- ganically modified silica nanoparticles accumulate in
nes (MCP-1 and MIP-2) [33, 84] and cytokines, such all organs, there were no inducted signs of organ tox-
as interleukins IL-1 [121], IL-8, IL-6 [32, 87] and icity [77]. Nishimori et al. [112] evaluating the acute
TNF-a [33, 121], CD54 and CD62E [32, 162]. Other toxicity of amorphous silica particles observed a sig-
researchers also reported that silica nanoparticles in- nificant hepatotoxicity. During chronic administration
duced apoptosis via JNK/p53-dependent, mitochon- nano-size materials cause liver fibrosis in mice [112].
drial pathways [84]. Chen et al. [29] suggested an ab-
sorption of SNPs in the nucleus generated aberrant
clusters of topoisomerase I and protein aggregates in
the nucleoplasm. The formation of nucleoplasmic ag-
Carbon nanomaterials
gregates impairs such nuclear functions as inhibiting
replication and transcription [29]. Yang and co-
workers implied that perturbation of intracellular free Carbon nanocarriers used in DDS are differentiated
calcium homeostasis may be responsible for cytotoxic into nanotubes (CNTs) and nanohorns (CNH).
effect of silica nanoparticles [169]. A recent work by CNTs are characterized by unique architecture
Zhao et al. [178] have demonstrated the effects of formed by rolling of single (SWNCTs single walled
nanoparticles (depending on the surface properties, carbon nanotubes) or multi (MWCNTs multi walled
structure and size) on human red blood cells (RBCs). carbon nanotubes) layers of graphite with an enor-
The uptake of large silica nanoparticles by RBCs mous surface area and an excellent electronic and
showed a strong local membrane deformation leading thermal conductivity [17]. Biocompatibility of nano-
to a spiculation of RBCs, internalization of the parti- tubes may be improved by chemical modification of
cles, and eventual hemolysis. On the contrary, adsorp- their surface [54]. Such adjustment can be imple-
tion of small particles occurred without affecting mented by covalent anchoring of PAMAM den-
membrane or morphology of RBCs [178]. drimers [176], amphiphilic diblock copolymers [45],

Pharmacological Reports, 2012, 64, 10201037 1027


or PEG layers [18] on CNTs surface or dispersion [6]. A comparison of these two paths of cisplatin in-
within a hyaluronic acid matrix [137]. Due to their corporation into nanohorns showed that nanoprecipi-
mechanical strength, SWCNTs have been used as tation is much more effective (almost 3-fold increase
a support to improve properties of other carriers, e.g., in the number of molecules entrapped in nanohorns)
polymeric or non-polymeric composites [137]. than adsorption [6].
There are three ways of drug immobilization in car- The toxicity of carbon nanomaterials also depends
bon nanocarriers, which are: encapsulation of a drug on their unique well-defined geometric structure [67].
in the carbon nanotube [11, 154], chemical adsorption The toxic potential of carbon nanotubes can result
on the surface or in the spaces between the nanotubes from the high length to diameter ratio and the toxicity
(by electrostatic, hydrophobic, p-p interactions and of the sole material, which is graphite. In addition,
hydrogen bonds) [31, 177], and attachment of active some impurities, such as residual metal and amor-
agents to functionalized carbon nanotubes (f-CNTs). phous carbon, contribute to the level increase of reac-
Encapsulation has the advantage over the two remain- tive oxygen species (ROS), thus, inducing the oxida-
ing methods as the drug is protected from degradation tive stress in cells [47]. Recent studies have pointed
during its transport to the cells and is released only in out the similarity in carcinogenic potential between
specific conditions [119]. The examples of drugs that CNT and asbestos. [120]. Carbon nanotubes have
were attached to CNTs are listed in Table 2. been shown to cause necrosis or apoptosis of macro-
Drug release from carbon nanotubes can be electri- phage cell lines and changes in cell morphology [67].
cally or chemically controlled. To prevent the un- Radomski et al. [127] studied the effects of engi-
wanted release of the drug, the open ends of CNTs neered carbon nanoparticles (MWCNT and SWCNT)
were sealed with polypyrrole (PPy) films [88]. Hom- on human platelet aggregation in vitro and rat vascu-
ing devices, i.e., folic acid [44] and epidermal growth lar thrombosis in vivo. Incubation of platelets with
factor [19], were attached to improve selectivity of carbon nanomaterials caused platelet aggregation
such drug delivery systems. with little or no granular release [127]. Incubation of
Nanohorns a type of the only single-wall nano- bronchial epithelial cells and keratinocytes with high
tubes exhibit similar properties to nanotubes [136]. doses of SWCNT resulted in oxidative stress, includ-
Their formation process does not require a metal cata- ing ROS generation, lipid peroxidation and mitochon-
lyst, thus, they can be easily prepared with very low drial dysfunction [133]. In vivo studies concerning
cost and are of high purity [136]. The immobilization intratracheal administration of nanotubes in rats
of drugs may rely on adsorption on nanohorns walls revealed the induction of change in lung structures,
[105] or nanoprecipitation of drugs with nanohorns such as granuloma formation and interstitial fibrosis

Tab. 2. Carbon nanotubes as DDS

Type of nanotubes Drug Method of immobilization Ref.


MWCNTs Cisplatin Encapsulation via capillary forces 154
f-CNTs Amphotericin B Conjugated to carbon nanotubes 123
SWCNTs Gemcitabine Encapsulation 11
MWNTs Epirubicin hydrochloride Adsorption 32
MWCNTs@poly(ethylene Doxorubicin Adsorption 45
glycol-b-propylene sulfide)
f-CNTs Sulfamethoxazole Adsorption 177
SWNTs-PL-PEG-NH Pt(IV) prodrug-FA Covalent amide linkages 44
SWNTs Cisplatin EGF Attachment to carbon nanotubes via amide 19
linkages
MWCNTs Dexamethasone Encapsulation 88

MWNTs multi walled nanotubes; f-CNTs functionalized carbon nanotubes; SWNTs-PL-PEG-NH amine-functionalized single-walled carbon
nanotubes

1028 Pharmacological Reports, 2012, 64, 10201037


Nanoparticles and drugs
Agnieszka Z. Wilczewska et al.

[80, 101]. Unmodified MWCNT caused pro-inflam- Therefore, designing magnetic drug delivery systems
matory response in keratinocytes cell lines, e.g., re- requires taking into consideration many factors, e.g.,
lease of cytokine interleukin 8 and formation of cyto- magnetic properties and size of particles, strength of mag-
plasmic vacuoles [98]. netic field, drug loading capacity, the place of accessibil-
ity of target tissue, or the rate of blood flow [24].
Depending on magnetic properties, MNPs can be
divided into pure metals (such as cobalt [96], nickel
Magnetic nanoparticles [69], manganese [132], and iron [143]), their alloys
and oxides. However, narrowing the area of MNPs
applications only to biomedicine reduces significantly
Magnetic nanoparticles exhibit a wide variety of at- the choice of magnetic material. Such a restriction re-
tributes, which make them highly promising carriers sults from the lack of knowledge of the negative ef-
for drug delivery. In particular, these are: easy han-
fects which the majority of these nanomaterials have
dling with the aid of an external magnetic field, the
on the human body.
possibility of using passive and active drug delivery
Iron oxide nanoparticles, due to the favorable fea-
strategies, the ability of visualization (MNPs are used
tures they exhibit, are the only type of MNPs ap-
in MRI), and enhanced uptake by the target tissue re-
proved for clinical use by Food and Drug Administra-
sulting in effective treatment at the therapeutically op-
tion. These attributes are: facile single step synthesis
timal doses [10].
However, in most of the cases where magnetic by alkaline co-precipitation of Fe + and Fe3+ [53],
nanocarriers have been used, difficulties in achieving chemical stability in physiological conditions [12]
these objectives appeared. It is most likely associated and possibility of chemical modification by coating
with inappropriate features of magnetic nanoparticles the iron oxide cores with various shells, i.e., golden
or inadequate magnet system. Magnetic nanoparti- [148], polymeric [34], silane [25], or dendrimeric
cles, for instance, tend to aggregate into larger clus- [114] (Fig. 3). In addition, iron oxides magnetite
ters loosing the specific properties connected with and maghemite occur naturally in human heart,
their small dimensions and making physical handling spleen and liver [57], which indicates their biocom-
difficult. In turn, magnetic force may not be strong patibility and non-toxicity at a physiological concen-
enough to overcome the force of blood flow and to tration. It is essential to estimate a safe upper limit of
accumulate magnetic drugs only at target site [110]. MNPs for biomedical use.

Fig. 3. Magnetic nanoparticles with various shells

Pharmacological Reports, 2012, 64, 10201037 1029


Connecting a drug with MNP may be achieved by involved in dissolving blood clots was covalently
covalent binding [53], electrostatic interactions [53], coupled to silanized [71] and chitosan-modified [28]
adsorption [168], or encapsulation process [166]. Tar- magnetic nanoparticles. The preliminary studies indi-
geting of drug-MNPs conjugates to diseased tissues cate that such conjugates can be useful in magneti-
(magnetic targeted drug delivery systems MTDDS), cally targeted lysis of in-stent thrombosis and can im-
depending on their size and surface chemistry, can be prove clinical aspects of thrombolytic therapy.
carried out by passive or active mechanism. Passive The influence of iron oxide NPs on cellular func-
targeting is a result of enhanced vascular permeability tion and toxicity has been investigated and reported.
and retention (EPR) of tumor tissues. Active strategy MNPs, depending on the way of their administration,
relies on the attraction of nanoparticle to the affected may interact with extracellular components or/and
site by using recognition ligands (e.g., antibodies) at- with the cell membranes of macrophages, endothelial
tached to the surface of MNPs and by handling of an cells, skin epithelium, and respiratory or gastrointesti-
external magnetic field [53]. nal tracts [35, 86, 89]. After inhalation, MNPs accu-
Therapeutic activity of diverse drugs incorporated mulate in the brain which indicates their ability to
into iron oxide nanocarriers have been tested and re- cross the blood-brain barrier. Mechanisms of inter-
ported (Tab. 3). MNPs have been examined for multi- nalization and overall biodistribution of MNPs are
tasking treatment as biosensors (diagnosis) and drug car- closely associated with their surface chemistry and
riers (therapy) simultaneously. Concomitant use of mag- hydrodynamic sizes [172]. Magnetic nanoparticles
netic resonance or magnetofluorescent imaging and can be quickly opsonized by plasma proteins, recog-
targeted therapy (via conjugation of targeting moieties) nized and subsequently removed from the blood-
can enhance effectiveness of cancer therapy [34]. stream by macrophages of the reticuloendothelial sys-
MNPs have been also tested as carriers for the treat- tem [138]. The greatest overall uptake of nanoparti-
ment of in-stent thrombosis. Traditional thrombolytic cles can be observed in liver and spleen [160].
therapy is associated with severe side effects, such as Investigations concerning the deformation of cells
hemorrhagic complications. In order to eliminate these upon their exposure to nanoparticles have revealed that
issues, a tissue plasminogen activator (tPA) a protein the toxicity of MNPs (at the same molarity) increases

Tab. 3. Magnetic nanoparticles as DDS

Drug Therapeutic activity Nanocarrier (core@shell) Ref.


Ciprofloxacin Anti-infective agents (antibiotic) Fe!O"@poly(vinyl alcohol)-g-poly(methyl 15
methacrylate)
Gemcitabine Antimetabolites, cancer chemotherapy Fe!O"@poly(ethylene glycol) 151
Doxorubicin Antineoplastic agent Fe!O"@gelatin 56
5-Fluorouracil Antimetabolites, anticancer drug Fe!O"@ethylcellulose 9
Daunorubicin Chemotherapeutic leukemia drug Fe!O" 166
Anti-b-HCG monoclonal Choriocarcinoma-specific gene vector Fe!O"@dextran 68
antibody
Cisplatin Chemotherapeutic drug Fe!O"@poly e-caprolactone 170
Paclitaxel Mitotic inhibitor used in cancer Fe!O"@poly[aniline-co-sodium N-(1-butyric acid) 65
chemotherapy
1,3-Bis(2-chloroethyl)-
1-nitrosourea (BCNU) Anti-cancer chemotherapy drug Fe!O"@poly[aniline-co-N-(1-butyric acid) aniline] 64
Tissue plasminogen activator, Fe!O"@tetraethyl orthosilicate 71
t-PA thrombolytic therapy Fe!O"@chitosan 28
Dopamine Catecholamine neurotransmitter, Fe!O"@silica (diatom) 86
Parkinsons disease

rd@ functionalization

1030 Pharmacological Reports, 2012, 64, 10201037


Nanoparticles and drugs
Agnieszka Z. Wilczewska et al.

in the following order: from nanobeads, to nanoworms oped and are under preclinical evaluation, only
and nanospheres [90]. MNPs coated with long-chain a handful of nanoparticle drugs are available on the
polymers are less cell toxic (slightly lower cell viability pharmaceutical market, e.g., liposomal conjugates:
in relation to the control probe in MTT assay) than Doxil (doxorubicin) or DaunoXome (daunorubi-
MNPs coated with shorter polymer chain of the same cin). It is due to the fact that nanoparticle based drug
type (with a significantly reduced viability) [58]. delivery systems do have a lot of drawbacks and limi-
Magnetic nanoparticles promote activation of tations. Some of them arise from scaling up problems.
phagocytotic and cytokine-release functions of macro- For instance, small size and large surface area of
phages [47]. Gas vesicles were observed in MNPs- nanoparticle-based targeting system can lead to an ag-
treated cells with increased granularity of the cells [89]. gregation, making physical handling difficult. Nano-
Magnetite nanoparticles have the potency of causing carrier-drug conjugates can be phagocytosed by cells
oxidative DNA lesions in cultured A549 cells (the hu- whereas their intracellular degradation may cause cy-
man lung epithelial cell line) [70]. They also cause totoxic effects. Other issues include low drug loading
transient increase of ALT (serum alanine aminotransfe-
capacity, low loading efficiency, and poor ability to
rase), AST (aspartate aminotransferase), and ALP (al-
control the size distribution of carriers. Furthermore,
kaline phosphatase) levels [66]. In several reports iron
there is a lack of technological methods, which will
oxide was found to accumulate in tissues but without
lead to nanodevices of approvable quality.
significant histological changes in vital organs [166].
Despite all the limitations and shortcomings, nano-
It is known that iron oxide NPs can induce oxida-
particle DDS which respond to slight changes in the
tive stress by excess of ROS production. The response
of defense elements leads to increased expression of local cellular environment have a potential to resolve
antioxidant enzymes, such as heme oxygenase 1 and many of the current drug delivery problems. How-
superoxide dismutase. This stage is followed by pro- ever, before the ongoing research will bring a clini-
inflammatory response, i.e., cytokine, chemokine, and cally useful drug delivery system, challenges which
matrix metalloproteinase (MMP) release, leading to include developing toxicity testing protocols, improv-
apoptosis and mutagenesis. An increase in the activity ing biocompatibility, drug loading, targeting, trans-
of MMP in the nervous system can enlarge BBB per- port and release, controlling interaction with biologi-
meability and cause neuronal damages. Oxidative cal barriers, detecting and monitoring exposure level
stress-induced cell injury and death may be avoided and assessing the impact on the environment have to
using appropriate dose of MNPs [107]. be met.
Due to a number of functional groups on the sur-
face of nanoparticles, the drug can be attached to the
carrier only in a stoichiometric ratio. The oxidative
Conclusion stress and inflammation in different cell types have
been often reported as toxic mechanisms of various
types of nanoparticles. Nanoparticles of diameter
Nanocarriers as drug delivery systems are designed to
10 nm can remain in cells and induct chronic inflam-
improve the pharmacological and therapeutic proper-
ties of conventional drugs. The incorporation of drug matory response and fibrosis of tissue. An additional
molecules into nanocarrier can protect a drug against problem is the lack of knowledge concerning the dis-
degradation as well as offers possibilities of targeting tribution of drug carriers and the unpredictability of
and controlled release. Due to small dimensions, the process. Thus, in our opinion, the magnetic tar-
nanocarriers are able to cross the blood-brain-barrier geted drug delivery system is one of the most attrac-
(BBB) and operate on cellular level. In comparison tive strategy target therapy. Magnetic nanoparticles
with the traditional form of drugs, nanocarrier-drug have their unique magnetic properties and they can be
conjugates are more effective and selective. They can attracted by magnetic fields, thus, acting as drug carri-
reduce the toxicity and other adverse side effects in ers in a target therapy. In addition, inorganic magnetic
normal tissues by accumulating drugs in target sites. nanoparticles containing the iron and gadolinium
In consequence, the required doses of drugs are lower. serve as an excellent contrast enhancing agents in
Although there are several nanoparticle-based MRI (approved by FDA Food and Drug Administra-
therapeutic agents which are currently being devel- tion).

Pharmacological Reports, 2012, 64, 10201037 1031


A real therapeutic breakthrough can be achieved 12. Asmatulu R, Zalich MA, Claus RO, Riffle J: Synthesis,
characterization and targeting of biodegradable magnetic
solely by carrying out painstaking studies in the field
nanocomposite particles by external magnetic fields.
of nano-therapy. Using nanosystems in therapies of J Magn Magn Mater, 2005, 292, 108119.
diseases may contribute to achieving an effective can- 13. Attama AA, Schicke BC, Paepenmller T, Mller-
cer treatment. Moreover, immobilization of homing Goymann CC: Solid lipid nanodispersions containing
devices, such as folic acid, epidermal growth factor or mixed lipid core and a polar heterolipid: characteriza-
antibodies, to the surface of nanoparticles, improves tion. Eur J Pharm Biopharm, 2007, 67, 4857.
14. Bai J, Li Y, Du J, Wang S, Zheng J, Yang O, Chen X:
selectivity of drug carriers. The key applications of One-pot synthesis of polyacrylamide-gold nanocompos-
nanoparticles in medicine are diagnosis and target ite. Mater Chem Phys, 2007, 106, 412415.
therapy, however, their wider use is still the future. 15. Bajpai AK, Gupta R: Magnetically mediated release of
ciprofloxacin from polyvinyl alcohol based superpara-
magnetic nanocomposites. J Mater Sci Mater Med, 2011,
22, 357369.
16. Balogh L, Swanson DR, Tomalia DA, Hagnauer GL,
References: McManus AT: Dendrimer-silver complexes and nano-
composites as antimicrobial agents. Nano Lett, 2001, 1,
1821.
1. Abdel-Mottaleb MMA, Neumann D, Lamprecht A:
17. Beg S, Rizwan M, Sheikh AM, Hasnain MS, Anwer K,
Lipid nanocapsules for dermal application: A compara-
Kohli K: Advancement in carbon nanotubes: basics, bio-
tive study of lipid-based versus polymer-based nanocar-
medical applications and toxicity. J Pharm Pharmacol,
riers. Eur J Pharm Biopharm, 2011, 79, 3642.
2011, 63, 141163.
2. Afergan E, Epstein H, Dahan R, Koroukhov N, Rohekar
K, Danenberg HD, Golomb G: Delivery of serotonin to 18. Bhirde AA, Patel S, Sousa AA, Patel V, Molinolo AA,
the brain by monocytes following phagocytosis of lipo- Ji Y, Leapman RD et al.: Distribution and clearance of
somes. J Control Release, 2008, 132, 8490. PEG-single-walled carbon nanotube cancer drug delivery
3. Agnihotri SA, Aminabhavi TM: Novel interpenetrating vehicles in mice. Nanomedicine, 2010, 5, 15351546.
network chitosan-poly(ethylene oxide-g-acrylamide)hy- 19. Bhirde AA, Patel V, Gavard J, Zhang G, Sousa AA,
drogel microspheres for the controlled release of cape- Masedunskas A, Leapman RD et al.: Targeted killing of
citabine. Int J Pharm, 2006, 324, 103115. cancer cells in vivo and in vitro with EGF-directed car-
4. Ahola MS, Silynoja ES, Raitavuo MH, Vaahtio MM, bon nanotube-based drug delivery. ACS Nano, 2009, 3,
Salonen JI, Yli-Urpo AU: In vitro release of heparin 307316.
from silica xerogels. Biomaterials, 2001, 22, 21632170. 20. Bilensoy E, Sarisozen C, Esendagl G, Dogan LA, Aktas
5. Ai J, Biazar E, Montazeri M, Majdi A, Aminifard S, Y, Sen M, Mangan AN: Intravesical cationic nanoparti-
Safari M, Akbari HR: Nanotoxicology and nanoparticle cles of chitosan and polycaprolactone for the delivery of
safety in biomedical designs. Int J Nanomedicine, 2011, Mitomycin C to bladder tumors. Int J Pharm, 2009, 371,
6, 11171127. 170176.
6. Ajima K, Murakami T, Mizoguchi Y, Tsuchida K, Ichi- 21. Biswas S, Dodwadkar NS, Deshpande PP, Torchilin VP:
hashi T, Iijima S, Yudasaka M: Enhancement of in vivo Liposomes loaded with paclitaxel and modified with
anticancer effects of cisplatin by incorporation inside single- novel triphenylphosphonium-PEG-PE conjugate possess
wall carbon nanohorns. ACS Nano, 2008, 2, 20572064. low toxicity, target mitochondria and demonstrate en-
7. Amato G: Silica-encapsulated efficient and stable si hanced antitumor effects in vitro and in vivo. J Control
quantum dots with high biocompatibility. Nanoscale Res Release, 2012, 159, 393402.
Lett, 2010, 5, 11561160. 22. Biswas S, Dodwadkar NS, Sawant RR, Torchilin VP:
8. Arayachukeat S, Wanichwecharungruang SP, Tree-Udom Development of the novel PEG-PE-based polymer for
T: Retinyl acetate-loaded nanoparticles: Dermal penetra- the reversible attachment of specific ligands to lipo-
tion and release of the retinyl acetale. Int J Pharm, 2011, somes: synthesis and in vitro characterization. Bioconjug
404, 281288. Chem, 2011, 22, 20052013.
9. Arias JL, Lpez-Viota M, Delgado AV, Ruiz MA: 23. Caminade AM, Laurent R, Majoral JP: Characterization
Iron/ethylcellulose (core/shell) nanoplatform loaded with of dendrimers. Adv Drug Deliv Rev, 2005, 57, 2130 2146.
5-fluorouracil for cancer targeting. Colloids Surf B Bio- 24. Cao Q, Han X, Li L: Enhancement of the efficiency of
interfaces, 2010, 77, 111116. magnetic targeting for drug delivery: Development and
10. Arruebo M, Fernndez-Pacheco R, Ibarra, MR, Santa- evaluation of magnet system. J Magn Magn Mater, 2011,
mara J: Magnetic nanoparticles for drug delivery. Nano 323, 19191924.
Today, 2007, 2, 2232. 25. Chang JH, Kang KH, Choi J, Jeong YK: High efficiency
11. Arsawang U, Saengsawang O, Rungrotmongkol T, protein separation with organosilane assembled silica
Sornmee P, Wittayanarakul K, Remsungnen T, coated magnetic nanoparticles. Superlattice Microst,
Hannongbua S: How do carbon nanotubes serve as 2008, 44, 442448.
carriers for gemcitabine transport in a drug delivery sys- 26. Chauhan AS, Diwan PV, Jain NK, Tomalia DA: Unex-
tem? J Mol Graph Model, 2011, 29, 591596. pected in vivo anti-inflammatory activity observed for

1032 Pharmacological Reports, 2012, 64, 10201037


Nanoparticles and drugs
Agnieszka Z. Wilczewska et al.

simple, surface functionalized poly(amidoamine) den- san nanoparticles for anti-HIV drug delivery applica-
drimers. Biomacromolecules, 2009, 10, 11951202. tions. Carbohydr Polym, 2010, 80, 833838.
27. Chauhan AS, Jain NK, Diwan PV: Pre-clinical and be- 44. Dhar S, Liu Z, Thomale J, Dai H, Lippard SJ: Targeted
havioural toxicity profile of PAMAM dendrimers in single-wall carbon nanotube-mediated Pt(IV) prodrug
mice. Proc R Soc A, 2010, 466, 15351550. delivery using folate as a homing device. J Am Chem
28. Chen JP, Yang PC, Ma YH, Wu T: Characterization of Soc, 2008, 27, 130, 1146711476.
chitosan magnetic nanoparticles for in situ delivery of 45. Di Crescenzo A, Velluto D, Hubbell JA, Fontana A:
tissue plasminogen activator. Carbohydr Polym, 2011, Biocompatible dispersions of carbon nanotubes: a poten-
84, 364372. tial tool for intracellular transport of anticancer drugs.
29. Chen M, von Mikecz A: Formation of nucleoplasmic Nanoscale, 2011, 3, 925928.
protein aggregates impairs nuclear function in response 46. Di Pasqua AJ, Wallner S, Kerwood DJ, Dabrowiak JC:
to SiO nanoparticles. Exp Cell Res, 2005, 305, 5162. Adsorption of the Pt(II) anticancer drug carboplatin by
30. Chen Z, Meng H, Xing G, Yuan H, Zhao F, Liu R, mesoporous silica. Chem Biodiv, 2009, 6, 13431349.
Chang X et al.: Age-related differences in pulmonary 47. Dobrovolskaia MA, McNeil SE: Immunological proper-
and cardiovascular responses to SiO nanoparticle inha- ties of engineered nanomaterials. Nat Nano, 2007, 2,
lation: nanotoxicity has susceptible population. Environ 469478.
Sci Technol, 2008, 42, 89858992. 48. dos Santos Giuberti C, de Oliveira Reis EC, Ribeiro
31. Chen Z, Pierre D, He H, Tan S, Pham-Huy C, Hong H, Rocha TG, Leite EA, Lacerda RG, Ramaldes GA,
Huang J: Adsorption behavior of epirubicin hydrochlo- de Oliveira MC: Study of the pilot production process
ride on carboxylated carbon nanotubes. Int J Pharm, of long-circulating and pH-sensitive liposomes contain-
2011, 28, 405, 153161. ing cisplatin. J Liposome Res, 2011, 21, 6069.
32. Cho M, Cho WS, Choi M, Kim SJ, Han BS, Kim SH, 49. Duncan R, Izzo L: Dendrimer biocompatibility and tox-
Kim HO et al.: The impact of size on tissue distribution icity. Adv Drug Deliv Rev, 2005, 57, 22152237.
and elimination by single intravenous injection of silica 50. Echeverra JC, Estella J, Barbera V, Musgo J, Garrido
nanoparticles. Toxicol Lett, 2009, 189, 177183. JJ: Synthesis and characterization of ultramicroporous
33. Cho WS, Choi M, Han BS, Cho M, Oh J, Park K, Kim silica xerogels. J Non-Cryst Solids, 2010, 356, 378382.
51. Eom HJ, Choi J: Oxidative stress of silica nanoparticles
SJ et al.: Inflammatory mediators induced by intratra-
in human bronchial epithelial cell, Beas-2B. Toxicol In
cheal instillation of ultrafine amorphous silica particles.
Vitro, 2009, 23, 13261332.
Toxicol Lett, 2007, 175, 2433.
52. Fidalgo A, Lopez TM , Ilharco LM: Wet solgel silica
34. Chomoucka J, Drbohlavova J, Huska D, Adam V, Kizek
matrices as delivery devices for phenytoin. J Sol-Gel Sci
R, Hubalem J: Magnetic nanoparticles and targeted drug
Technol, 2009, 49, 320328.
delivering. Pharm Res, 2010, 62, 144149.
53. Figuerola A, Di Corato R, Manna L, Pellegrino T: From
35. Cole AJ, Yang VC, David AE: Cancer theranostics: the
iron oxide nanoparticles towards advanced iron-based in-
rise of targeted magnetic nanoparticles. Trends Biotech-
organic materials designed for biomedical applications.
nol, 2011, 29, 323332.
Pharmacol Res, 2010, 62, 126143.
36. Czarnobaj K: Preparation and characterization of silica xe- 54. Foldvari M, Bagonluri M: Carbon nanotubes as func-
rogels as carriers for drugs. Drug Deliv, 2008, 15, 485492. tional excipients for nanomedicines: I. Pharmaceutical
37. Czarnobaj K, Czarnobaj J: Sol-gel processed porous sil- properties. Nanomedicine, 2008, 4, 173182.
ica carriers for the controlled release of diclofenac dieth- 55. Fritzen-Garcia MB, Zanetti-Ramos BG, Schweitzer de
ylamine. J Biomed Mater Res B Appl Biomater, 2008 Oliveira C, Soldi V, Pasa AA, Creczynski-Pasa TB:
87, 114120. Atomic force microscopy imaging of polyurethane nano-
38. Czarnobaj K, Lukasiak J: In vitro release of cisplatin particles onto different solid. Mater Sci Eng C, 2009, 29,
from sol-gel processed organically modified silica xero- 405409.
gels. J Mater Sci Mate Med, 2007, 18, 20412044. 56. Gaihre B, Khil MS, Lee DR, Kim HY: Gelatin-coated
39. Czarnobaj K, Sawicki W: The sol-gel prepared magnetic iron oxide nanoparticles as carrier system: drug
SiO -CaO-P O# composites doped with metronidazole loading and in vitro drug release study. Int J Pharm,
for application in local delivery systems. Pharm Dev 2009, 365, 180189.
Technol, 2011, (doi: 10.3109/10837450.2011.572894). 57. Grassi-Schultheiss PP, Heller F, Dobson J: Analysis of
40. DEmanuele A, Attwood D: Dendrimer-drug interac- magnetic material in the human heart, spleen and liver.
tions. Adv Drug Deliv Rev, 2005, 57, 21472162. Biometals, 1997, 10, 351355.
41. Daemen T, Hofstede G, Ten Kate MT, Bakker-Woudenberg 58. Hafeli UO, Riffle JS, Harris-Shekhawat L, Carmichael-
IA, Scherphof GL: Liposomal doxorubicin-induced tox- Baranauskas A, Mark F, Dailey JP, Bardenstein D: Cell
icity: depletion and impairment of phagocytic activity of uptake and in vitro toxicity of magnetic nanoparticles suit-
liver macrophages. Int J Cancer, 1995, 61, 666671. able for drug delivery. Mol Pharm, 2009, 6, 14171428.
42. des Rieux A, Fievez V, Garinot M, Schneider YJ, 59. Han L, Huang R, Liu S, Huang S, Jiang Ch: Peptide-
Prat V: Nanoparticles as potential oral delivery systems conjugated PAMAM for targeted doxorubicin delivery to
of proteins and vaccines: a mechanistic approach. J Con- transferrin receptor overexpressed tumors. Mol Pharm,
trol Release, 2006,116, 127. 2010, 7, 21562165.
43. Dev A, Binulal NS, Anita A, Nair SV, Furuike T, Tamura 60. He Q, Gao Y, Zhang L, Zhang Z, Gao F, Ji X, Li Y,
H, Jayakumar R: Preparation of poly(lactic acid)/chito- Shi J: A pH-responsive mesoporous silica nanoparticles-

Pharmacological Reports, 2012, 64, 10201037 1033


based multi-drug delivery system for overcoming multi- charge and fluorescent labeling. Pharm Res, 2006, 23,
drug resistance. Biomaterials, 2011, 32, 77117720. 28182826.
61. Heiden TCK, Dengler E, Kao WJ, Heideman W, Peter- 75. Kojima C, Regino C, Umeda Y, Kobayashi H, Kono K:
son DE: Developmental toxicity of low generation Influence of dendrimer generation and polyethylene gly-
PAMAM dendrimers in zebrafish. Toxicol Appl Pharma- col length on the biodistribution of PEGylated den-
col, 2007, 225, 7079. drimers. Int J Pharm, 2010, 383, 293296.
62. Hocine O, Gary-Bobo M, Brevet D, Maynadier M, 76. Kovacevic A, Savic S, Vuleta G, Mller RH, Keck CM:
Fontanel S, Raehm L, Richeter S et al.: Silicalites and Polyhydroxy surfactants for the formulation of lipid
mesoporous silica nanoparticles for photodynamic nanoparticles (SLN and NLC): Effects on size, physical
therapy. Int J Pharm, 2010, 402, 221230. stability and particle matrix structure. Int J Pharm, 2011,
63. Hong S, Bielinska AU, Mecke A, Keszler B, Beals JL, 406, 163172.
Shi X, Balogh L et al.: Interaction of poly(amidoamine) 77. Kumar R, Roy I, Ohulchanskky TY, Vathy LA, Bergey
dendrimers with supported lipid bilayers and cells: hole EJ, Sajjad M, Prasad PN: In vivo biodistribution and
formation and the relation to transport. Bioconjug Chem, clearance studies using multimodal organically modified
2004, 15, 774782. silica nanoparticles. ACS Nano, 2010, 4, 699708.
64. Hua MY, Liu HL, Yang HW, Chen PY, Tsai RY, Huang 78. Kumari A, Yadav SK, Yadav SC: Biodegradable poly-
CY, Tseng IC et al.: The effectiveness of a magnetic meric nanoparticles based drug delivery systems. Col-
nanoparticle-based delivery system for BCNU in the loids Surf B Biointerfaces, 2010, 75, 118.
treatment of gliomas. Biomaterials, 2011, 32, 516527. 79. Lai J, Lu Y, Yin Z, Hu F, Wu W: Pharmacokinetics and
65. Hua MY, Yang HW, Chuang CK, Tsai RY, Chen WJ, enhanced oral bioavailability in beagle dogs of cyclo-
Chuang KL, Chang YH et al.: Magnetic-nanoparticle- sporine A encapsulated in glyceryl monooleate/polox-
modified paclitaxel for targeted therapy for prostate can- amer 407 cubic nanoparticles. Int J Nanomedicine, 2010,
cer. Biomaterials, 2010, 31, 73557363. 5, 1323.
80. Lam CW, James JT, McCluskey R, Hunter RL: Pulmo-
66. Jain TK, Reddy MK, Morales MA, Leslie-Pelecky DL,
nary toxicity of single-wall carbon nanotubes in mice 7
Labhasetwar V: Biodistribution, clearance, and biocom-
and 90 days after intratracheal instillation. Toxicol Sci,
patibility of iron oxide magnetic nanoparticles in rats.
2004, 77, 126134.
Mol Pharm, 2007, 5, 316327.
81. Li Z, Su K, Cheng B, Deng Y: Organically modified
67. Jia G, Wang H, Yan L, Wang X, Pei R, Yan T, Zhao Y,
MCM-type material preparation and its usage in con-
Guo X: Cytotoxicity of carbon nanomaterials: single-
trolled amoxicillin delivery. J Colloid Interface Sci,
wall nanotube, multi-wall nanotube, and fullerene. Envi-
2010, 342, 607 613.
ron Sci Technol, 2005, 39, 13781383.
82. Lin W, Huang YW, Zhou XD, Ma Y: In vitro toxicity of
68. Jingting C, Huining L, Yi Z: Preparation and characteri-
silica nanoparticles in human lung cancer cells. Toxicol
zation of magnetic nanoparticles containing
Appl Pharmacol, 2006, 217, 252259.
Fe!O"-dextran-anti-b-human chorionic gonadotropin,
83. Liu J, Gong T, Fu H, Wang C, Wang X, Chen Q, Zhang Q,
a new generation choriocarcinoma-specific gene vector. He Q, Zhang Z: Solid lipid nanoparticles for pulmonary
Int J Nanomedicine, 2011, 6, 285294. delivery of insulin, Int J Pharm, 2008, 356, 333344.
69. Kale SN, Jadhav AD, Verma S, Koppikar SJ, Kaul- 84. Liu X, Sun J: Endothelial cells dysfunction induced by
Ghanekar R, Dhole SD, Ogale SB: Characterization of silica nanoparticles through oxidative stress via JNK/P53
biocompatible NiCo O" nanoparticles for applications in and NF-kB pathways. Biomaterials, 2010, 31, 8198 8209.
hyperthermia and drug delivery. Nanomedicine, 2012, 8, 85. Lope AL, Reins RY, McDermott AM, Trautner BW,
452459. Cai C: Antibacterial activity and cytotoxicity of
70. Karlsson HL, Cronholm P, Gustafsson J, Moller L: PEGylated poly(amidoamine) dendrimers. Mol Biosyst,
Copper oxide nanoparticles are highly toxic: a compari- 2009, 5, 1148 1156.
son between metal oxide nanoparticles and carbon nano- 86. Losic D, Yu Y, Aw MS, Simovic S, Thierry B, Addai-
tubes. Chem Res Toxicol, 2008, 21, 17261732. Mensah J: Surface functionalisation of diatoms with do-
71. Kempe H, Kempe M: The use of magnetite nanoparticles pamine modified iron-oxide nanoparticles: toward mag-
for implant-assisted magnetic drug targeting in throm- netically guided drug microcarriers with biologically de-
bolytic therapy. Biomaterials, 2010, 31, 94999510. rived morphologies. Chem Commun (Camb), 2010, 46,
72. Kim JK, Choi SH, Kim CO, Park JS, Ahn WS, Kim CK: 63236325.
Enhancement of polyethylene glycol (PEG)-modified 87. Luo G, Yu X, Jin C, Yang F, Fu D, Long J, Xu J et al.:
cationic liposome-mediated gene deliveries: effects on LyP-1-conjugated nanoparticles for targeting drug deliv-
serum stability and transfection efficiency. J Pharm Phar- ery to lymphatic metastatic tumors. Int J Pharm, 2010,
macol, 2003, 55, 453460. 385, 150156.
73. Kim TW, Slowing II, Chung PW, Lin VS: Ordered 88. Luo X, Matranga C, Tan S, Alba N, Cui XT: Carbon
mesoporous polymer-silica hybrid nanoparticles as vehi- nanotube nanoreservior for controlled release of anti-
cles for the intracellular controlled release of macro- inflammatory dexamethasone. Biomaterials, 2011, 32,
molecules. ACS Nano, 2011, 5, 360366. 63166323.
74. Kitchens KM, Kolhatkar RB, Swaan PW, Eddington ND, 89. Mahmoudi M, Simchi A, Imani M, Shokrgozar MA,
Ghandehari H.: Transport of poly(amidoamine) den- Milani AS, Hafeli U, Stroeve P: A new approach for
drimers across Caco-2 cell monolayers: influence of size, the in vitro identification of the cytotoxicity of super-

1034 Pharmacological Reports, 2012, 64, 10201037


Nanoparticles and drugs
Agnieszka Z. Wilczewska et al.

paramagnetic iron oxide nanoparticles. Colloids Surf B 106. Nanjwade BK, Singh J, Parikh KA, Manvi FV: Prepara-
Biointerfaces, 2010, 75, 300309. tion and evaluation of carboplatin biodegradable poly-
90. Mahmoudi M, Simchi A, Milani AS, Stroeve P: Cell tox- meric nanoparticles. Int J Pharm, 2010, 385, 176180.
icity of superparamagnetic iron oxide nanoparticles. 107. Naqvi S, Samim M, Abdin MZ, Ahmed FJ, Maitra AN,
J Colloid Interface Sci, 2009, 336, 510518. Prashant CK, Dinda AK: Concentration-dependent toxic-
91. Mainardes RM, Khalil NM, Gremio MPD: Intranasal ity of iron oxide nanoparticles mediated by increased
delivery of zidovudine by PLA and PLAPEG blend oxidative stress. Int J Nanomedicine, 2010, 5, 983989.
nanoparticles. Int J Pharm, 2010, 395, 266271. 108. Natali S, Mijovic J: Dendrimers as drug carriers: dynam-
92. Malik N, Evagorou EG, Duncan R: Dendrimer-platinate: ics of PEGylated and methotrexate-loaded dendrimers in
a novel approach to cancer chemotherapy. Anti-Cancer aqueous solution. Macromolecules, 2010, 43, 30113017.
Drugs Des, 1999, 10, 767776. 109. Nayak AP, Tiyaboonchai W, Patankar S, Madhusudhan
93. Mao HQ, Roy K, Troung-Le VL, Janes KA, Lin KY, B, Souto EB: Curcuminoids-loaded lipid nanoparticles:
Wang J, August JT, Leong KW: Chitosan-DNA nanopar- novel approach towards malaria treatment. Colloids
ticles as gene carriers: synthesis, characterization and Surf B Biointerfaces, 2010, 81, 263273.
transfection efficiency. J Control Release, 2001, 70, 110. Neuberger T, Schopf B, Hofmann H, Hofmann M, von
399421. Rechenberg B: Superpara-magnetic nanoparticles for
94. Martinem A, Iglesias I, Lozano R, Teijon JM, Blanco biomedical applications: possibilities and limitations of
MD: Synthesis and characterization of thiolated a new drug delivery system. J Magn Magn Mater, 2005,
alginate-albumin nanoparticles stabilized by disulfide 293, 483496.
bonds. Evaluation as drug delivery systems. Carbohydr 111. Nevozhay D, Kaska U, Budzyska R, Boratyski J:
Polym, 2011, 83, 13111321. Current status of research on conjugates and related drug
95. Maver U, Godec A, Bele M, Planinek O, Gaberek M, delivery systems in the treatment of cancer and other dis-
Sri S, Jamnik J: Novel hybrid silica xerogels for stabi- eases (Polish). Postepy Hig Med Dosw, 2007, 61, 350360.
lization and controlled release of drug. Int J Pharm, 112. Nishimori H, Kondoh M, Isoda K, Tsunoda S, Tsutsumi
2007, 330,164174. Y, Yagi K: Silica nanoparticles as hepatotoxicants. Eur
96. Meng X, Seton HC, Lu le T, Prior IA, Thanh NT, Song J Pharm Biopharm, 2009, 72, 496501.
B: Magnetic CoPt nanoparticles as MRI contrast agent 113. Paavola A, Kilpelinen I, Yliruusi J, Rosenberg P: Con-
for transplanted neural stem cells detection. Nanoscale, trolled release injectable liposomal gel of ibuprofen for
2011, 3, 977984. epidural analgesia. Int J Pharm, 2000, 199, 8593.
97. Menjoge AR, Kannan RM, Tomalia DA: Dendrimer- 114. Pan BF, Gao F, Gu HC: Dendrimer modified magnetite
based drug and imaging conjugates: design considera- nanoparticles for protein immobilization. J Colloid Inter-
tions for nanomedical applications. Drug Discov Today, face Sci, 2005, 284, 16.
2010, 15, 171187. 115. Pandey R, Ahmad Z, Sharma S, Khuller GK: Nano-
98. Monteiro-Riviere NA, Nemanich RJ, Inman AO, Wang encapsulation of azole antifungals: Potential applications
YY, Riviere JE: Multi-walled carbon nanotube interac- to improve oral drug delivery. Int J Pharm, 2005, 301,
tions with human epidermal keratinocytes. Toxicol Lett, 268276.
2005, 155, 377384. 116. Panyam J, Labhasetwar V: Biodegradable nanoparticles
99. Mora-Huertas CE, Fessi H, Elaissari A: Polymer-based for drug and gene delivery to cells and tissue. Adv Drug
nanocapsules for drug delivery. Int J Pharm, 2010, 385, Deliv Rev, 2003, 55, 329347.
113142. 117. Park EJ, Park K: Oxidative stress and pro-inflammatory
100. Muchow M, Maincent P, Mller RH: Lipid nanoparticles responses induced by silica nanoparticles in vivo and in
with a solid matrix (SLN, NLC LDC) for oral drug deliv- vitro. Toxicol Lett, 2009, 184, 1825.
ery. Drug Dev Ind Pharm, 2008, 34, 13941405. 118. Park J, Fong PM, Lu J, Russell KS, Booth KJ, Saltzman
101. Muller J, Huaux F, Moreau N, Misson P, Heilier JF, De- WM, Fahmy TM: PEGylated PLGA nanoparticles for
los M, Arras M et al.: Respiratory toxicity of multi-wall the improved delivery of doxorubicin. Nanomedicine,
carbon nanotubes. Toxicol Appl Pharmacol, 2005, 207, 2009, 5, 410-418.
221231. 119. Perry JL, Martin CR, Stewart JD: Drug-delivery strate-
102. Mller RH, Keck CM: Challenges and solutions for the gies by using template-synthesized nanotubes. Chemis-
delivery of biotech drugs a review of drug nanocrystal try, 2011, 17, 62966302.
technology and lipid nanoparticles. J Biotechnol, 2004, 120. Poland CA, Duffin R, Kinloch I, Maynard A, Wallace
113, 151170. WA, Seaton A, Stone V et al.: Carbon nanotubes intro-
103. Mller RH, Petersen RD, Hommoss A, Pardeike J: duced into the abdominal cavity of mice show asbestos-
Nanostructured lipid carriers (NLC) in cosmetic dermal like pathogenicity in a pilot study. Nat Nanotechnol,
products. Adv Drug Deliv Rev, 2007, 59, 522530. 2008, 3, 423428.
104. Mller RH, Radtke M, Wissing SA: Nanostructured lipid 121. Popovici RF, Seftel EM, Mihai GD, Popovici E, Voicu
matrices for improved microencapsulation of drugs, Int VA: Controlled drug delivery system based on ordered
J Pharm, 2002, 242, 121128. mesoporous silica matrices of captopril as angiotensin-
105. Murakami T, Sawada H, Tamura G, Yudasaka M, Iijima converting enzyme inhibitor drug. J Pharm Sci, 2011,
S, Tsuchida K: Water-dispersed single-wall carbon nano- 100, 704714.
horns as drug carriers for local cancer chemotherapy. 122. Prajapati RN, Tekade RK, Gupta U, Gajbhiye V, Jain
Nanomedicine, 2008, 3, 453463. NK: Dendimer-mediated solubilization, formulation de-

Pharmacological Reports, 2012, 64, 10201037 1035


velopment and in vitro-in vivo assessment of piroxicam. 138. Shubayev VI, Pisanic TR 2nd, Jin S: Magnetic nanopar-
Mol Pharm, 2009, 6, 940950. ticles for theragnostics. Adv Drug Deliv Rev, 2009, 61,
123. Prajapati VK, Awasthi K, Gautam S, Yadav TP, Rai M, 467477.
Srivastava ON, Sundar S et al.: Targeted killing of Leish- 139. Silva R, Ferreira H, Cavaco-Paulo A: Sonoproduction
mania donovani in vivo and in vitro with amphotericin B of liposomes and protein particles as templates for deliv-
attached to functionalized carbon nanotubes. J Antimi- ery purposes. Biomacromolecules, 2011, 12, 33533368.
crob Chemother, 2011, 66, 874 879. 140. Singh P, Gupta U, Asthana A, Jain NK: Folate and
124. Prokopowicz M: Synthesis and in vitro characterization Folate-PEG-PAMAM dendrimers: synthesis, characteri-
of freeze-dried doxorubicin-loaded silica xerogels. zation, and targeted anticancer drug delivery potential
J Sol-Gel Sci Technol, 2010, 53, 525533. in tumor bearing mice. Bioconjugate Chem, 2008, 19,
125. Puglia C, Blasi P, Rizza L, Schoubben A, Bonina F, 22392252.
Rossi C, Ricci M: Lipid nanoparticles for prolonged 141. Slowing I, Trewyn BG, Giri S, Lin VS-Y: Mesoporous
topical delivery: an in vitro and in vivo investigation. silica nanoparticles for drug delivery and biosensing ap-
Int J Pharm, 2008, 357, 295304. plications. Adv Funct Mater, 2007, 17, 12251236.
126. Quintanar-Guerrero D, Ganem-Quintanar A, Nava- 142. Slowing I, Vivero-Escoto JL, Wu Ch-W, Lin VS-Y:
Arzaluz MG, Pin-Segundo E: Silica xerogels as phar- Mesoporous silica nanoparticles as controlled release
maceutical drug carriers. Expert Opin Drug Deliv, 2009, drug delivery and gene transfection carriers. Adv Drug
6, 485498. Deliv Rev, 2008, 60, 12781288.
127. Radomski A, Jurasz P, Alonso-Escolano D, Drews M, 143. Smolensky ED, Park HY, Berqu TS, Pierre VC: Surface
Morandi M, Malinski T, Radomski MW: Nanoparticle- functionalization of magnetic iron oxide nanoparticles
induced platelet aggregation and vascular thrombosis. for MRI applications - effect of anchoring group and
Br J Pharmacol, 2005, 146, 882893. ligand exchange protocol. Contrast Media Mol Imaging,
128. Rejinold NS, Chennazhi KP, Nair SV, Tamura H, 2011, 6, 189199.
Jayakumar R: Biodegradable and thermo-sensitive 144. Sunderland CJ, Steiert M, Talmadge JE, Derfus AM,
chitosan-g-poly(N-vinylcaprolactam) nanoparticles as Barry SE: Targeted nanoparticles for detecting and treat-
a 5-fluorouracil carrier. Carbohydr Polym, 2011, 83, ing cancer. Drug Dev Res, 2006, 67, 7093.
776786. 145. Suri SS, Fenniri H, Singh B: Nanotechnology-based drug
129. Roberts JC, Bhalgat MK, Zera RT: Preliminary biologi- delivery systems. J Occup Med Toxicol, 2007, 2, 16.
cal evaluation of polyamidoamine (PAMAM) Starburst 146. Svenson S, Tomalia DA: Dendrimers in biomedical ap-
dendrimers. J Biomed Mater Res, 1996, 30, 5365. plications reflections on the field. Adv Drug Deliv Rev,
130. Safdar A, Ma J, Saliba F, Dupont B, Wingard JR, 2005, 57, 2106 2129.
Hachem RY, Mattiuzzi GN et al.: Drug-induced nephro- 147. Sznitowska M, Gajewska M, Janicki S, Radwanska A,
toxicity caused by amphotericin B lipid complex and li- Lukowski G: Bioavailability of diazepam from
posomal amphotericin B: a review and meta-analysis. aqueous-organic solution, submicron emulsion and solid
Medicine (Baltimore), 2010, 89, 236244. lipid nanoparticles after rectal administration in rabbits.
131. Saraog GK, Gupta P, Gupta UD, Jain NK, Agrawal GP: Eur J Pharm Biopharm, 2001, 52, 159163.
Gelatin nanocarriers as potential vectors for effective 148. Tamer U, Gundogdu Y, Boyaci IH. Pekmez K: Synthesis
management of tuberculosis. Int J Pharm, 2010, 385, of magnetic core-shell Fe!O" Au, nanoparticles for
143149. biomolecule immobilization and detection. J Nanopart
132. Sayed FN, Jayakumar OD, Sudakar C, Naik R, Res, 2010, 12, 11871196.
Tyagi AK: Possible weak ferromagnetism in pure and 149. Tanis I, Karatasos K: Association of a weakly acidic
M (Mn, Cu, Co, Fe and Tb) doped NiGa O" nanoparti- anti-inflammatory drug (Ibuprofen) with a poly(ami-
cles. J Nanosci Nanotechnol, 2011, 11, 33633369. doamine) dendrimer as studied by molecular dynamics
133. Sayes CM, Liang F, Hudson JL, Mendez J, Guo W, simulations. J Phys Chem B, 2009, 113, 1098410993.
Beach JM, Moore VC et al.: Functionalization density 150. Tomalia DA: Birth of a new macromolecular architec-
dependence of single-walled carbon nanotubes cytotox- ture: dendrimers as quantized building blocks for
icity in vitro. Toxicol Lett, 2006, 161, 135142. nanoscale synthetic organic chemistry. Prog Polym Sci,
134. Shah N, Steptoe RJ, Parekh HS: Low-generation asym- 2005, 30, 294324.
metric dendrimers exhibit minimal toxicity and effec- 151. Tong Q, Li H, Li W, Chen H, Shu X, Lu X, Wang G:
tively complex DANN. J Pept Sci, 2011, 17, 470478. In vitro and in vivo anti-tumor effects of gemcitabine
135. Shi L, Fleming CJ, Riechers SL, Yin N-N, Luo J, loaded with a new drug delivery system. J Nanosci
Lam KS, Liu GY: High-resolution imaging of dendrimers Nanotechnol, 2011, 11, 36513658.
used in drug delivery via scanning probe microscopy. 152. Torchilin V: Multifunctional Pharmaceutical Nanocarri-
J Drug Deliv, 2011, (doi:10.1155/2011/254095). ers, Springer Science + Business Media, LLC, NY, 2008.
136. Shiba K, Yudasaka M, Iijima S: Carbon nanohorns as 153. Tosh DK, Yoo LS, Chinn M, Hong K, Kilbey MS, Bar-
a novel drug carrier. Nihon Rinsho, 2006, 64, 239246. rett MO, Fricks IP et al.: Polyamidoamine (PAMAM)
137. Shin US, Yoon IK, Lee GS, Jang WC, Knowles JC, dendrimer conjugates of clickable agonists of the A!
Kim HW: Carbon nanotubes in nanocomposites and hy- adenosine receptor and coactivation of the P2Y" recep-
brids with hydroxyapatite for bone replacements. J Tis- tor by a tethered nucleotide. Bioconjug Chem, 2010, 21,
sue Eng, 2011, (doi:10.4061/2011/674287). 372-384.

1036 Pharmacological Reports, 2012, 64, 10201037


Nanoparticles and drugs
Agnieszka Z. Wilczewska et al.

154. Tripisciano C, Costa S, Kalenczuk RJ, Borowiak-Palen 168. Yallapu MM, Othman SF, Curtis ET, Gupta BK, Jaggi
E: Cisplatin filled multiwalled carbon nanotubes a novel M, Chauhan SC: Multi-functional magnetic nanoparti-
molecular hybrid of anticancer drug container. Eur Phys cles for magnetic resonance imaging and cancer therapy.
J B, 2010, 75, 141146. Biomaterials, 2011, 32, 18901905.
155. Turkova A, Roilides E, Sharland M: Amphotericin B in 169. Yang H, Wu Q, Tang M, Kong L, Lu Z: Cell membrane
neonates: deoxycholate or lipid formulation as first-line injury induced by silica nanoparticles in mouse macro-
therapy - is there a right choice? Curr Opin Infect Dis, phage. J Biomed Nanotechnol, 2009, 5, 528535.
2011, 24, 163171. 170. Yang J, Park SB, Yoon HG, Huh YM, Haam S: Prepara-
156. Turos E, Shim JY, Wang Y, Greenhalgh K, Reddy GS, tion of poly e-caprolactone nanoparticles containing
Dickey S, Lim DV: Antibiotic-conjugated polyacrylate magnetite for magnetic drug carrier. Int J Pharm, 2006,
nanoparticles: New opportunities for development of 324, 185190.
anti-MRSA agents. Bioorg Med Chem Lett, 2007, 17, 171. Yeung CK, Shek SY, Yu CS, Kono T, Chan HH: Lipo-
5356. some-encapsulated 0.5% 5-aminolevulinic acid with in-
157. ner M, Yener G: Importance of solid lipid nanoparticles tense pulsed light for the treatment of inflammatory facial
(SLN) in various administration routes and future per- acne: a pilot study. Dermatol Surg, 2011, 37, 450459.
spectives. Int J Nanomedicine, 2007, 2, 289300. 172. Yoo JW, Chambers E, Mitragotri S: Factors that control
158. van den Hoven JM, Van Tomme SR, Metselaar JM, the circulation time of nanoparticles in blood: chal-
Nuijen B, Beijnen JH, Storm G: Liposomal drug formu- lenges, solutions and future prospects. Curr Pharm Des,
lations in the treatment of rheumatoid arthritis. Mol 2010, 16, 22982307.
Pharm, 2011, 8, 10021015. 173. Yu H, Nie Y, Dohmen Ch, Li Y, Wagner E: Epidermal
159. Waite CL, Roth ChM: PAMAM-RGD conjugates en- growth factor-PEG functionalized PAMAM-penta-
hance siRNA delivery through a multicellular spheroid ethylenehexamine dendron for targeted gene delivery
model of malignant glioma. Bioconjug Chem, 2009, 20, produced by click chemistry. Biomacromolecules, 2011,
19081916. 12, 20392047.
160. Wang J, Chen Y, Chen B, Ding J, Xia G, Gao C,
174. Yu KO, Grabinski CM, Schrand AM, Murdock RC,
Cheng J et al.: Pharmacokinetic parameters and tissue
Wang W, Gu B, Schlager JJ et al.: Toxicity of amorphous
distribution of magnetic Fe!O" nanoparticles in mice.
silica nanoparticles in mouse keratinocytes. J Nanopart
Int J Nanomedicine, 2010, 5, 861866.
Res, 2009, 11, 1524.
161. Wngler C, Moldenhauer G, Eisenhut M, Haberkorn U,
175. Yukihara M, Ito K, Tanoue O, Goto K, Matsushita T,
Mier W: Antibody-dendrimer conjugates: the number,
Matsumoto Y, Masuda M et al.: Effective drug delivery
not the size of the dendrimers, determines the imunore-
system for duchenne muscular dystrophy using hybrid
activity. Bioconjug Chem, 2008, 19, 813820.
liposomes including gentamicin along with reduced tox-
162. Waters KM, Masiello LM, Zangar RC, Tarasevich BJ,
icity. Biol Pharm Bull, 2011, 34, 712716.
Karin NJ, Quesenberry RD, Bandyopadhyay S et al.:
176. Zhang B, Chen Q, Tang H, Xie Q, Ma M, Tan L, Zhang
Macrophage responses to silica nanoparticles are highly
Y, Yao S: Characterization of and biomolecule immobili-
conserved across particle sizes. Toxicol Sci, 2009, 107,
zation on the biocompatible multi-walled carbon nano-
553569.
163. Wei L, Hu N, Zhang Y: Synthesis of polymer-mesoporous tubes generated by functionalization with polyami-
silica nanocomposites. Materials, 2010, 3, 40664079. doamine dendrimers. Colloids Surf B Biointerfaces,
164. Wilson B, Samanta MK, Santhi K, Kumar KP, 2010, 80, 1825.
Ramasamy M, Suresh B: Chitosan nanoparticles as 177. Zhang D, Pan B, Wu M, Wang B, Zhang H, Peng H,
a new delivery system for the anti-Alzheimer drug Wu D, Ning P: Adsorption of sulfamethoxazole on func-
tacrine. Nanomedicine, 2010, 6, 144152. tionalized carbon nanotubes as affected by cations and
165. Wissing SA, Kayser O, Mller RH: Solid lipid nanopar- anions. Environ Pollut, 2011, 159, 26162621.
tic les for parenteral drug delivery. Adv Drug Deliv Rev, 178. Zhao Y, Sun X, Zhang G, Trewyn BG, Slowing II,
2004, 56, 12571272. Lin VS: Interaction of mesoporous silica nanoparticles
166. Wu W, Chen B, Cheng J, Wang J, Xu W, Liu L, Xia G with human red blood cell membranes: size and surface
et al.: Biocompatibility of Fe!O"/DNR magnetic nano- effects. ACS Nano, 2011, 5, 13661375.
particles in the treatment of hematologic malignancies.
Int J Nanomedicine, 2010, 5, 10791084.
167. Xiong F, Mi Z, Gu N: Cationic liposomes as gene deliv-
ery system: transfection efficiency and new application. Received: November 24, 2011; in the revised form: May 7, 2012;
Pharmazie, 2011, 66, 158164. accepted: May 22, 2012.

Pharmacological Reports, 2012, 64, 10201037 1037

S-ar putea să vă placă și