Sunteți pe pagina 1din 10

International Journal of Pharmaceutics 516 (2017) 6170

Contents lists available at ScienceDirect

International Journal of Pharmaceutics


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

Folic acid-decorated and PEGylated PLGA nanoparticles for improving


the antitumour activity of 5-uorouracil
Mazen M. El-Hammadia,b , ngel V. Delgadoc, Consolacin Melguizod,e, Jos C. Pradosd,e,
Jos L. Ariasa,d,e,*
a
Department of Pharmacy and Pharmaceutical Technology, Faculty of Pharmacy, University of Granada, Spain
b
Department of Pharmaceutics and Pharmaceutical Technology, Faculty of Pharmacy, University of Damascus, Syria
c
Department of Applied Physics, Faculty of Sciences, University of Granada, Spain
d
Institute of Biopathology and Regenerative Medicine (IBIMER), University of Granada, Spain
e
Biosanitary Institute of Granada (ibs.GRANADA), Andalusian Health Service (SAS), University of Granada, Spain

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

Article history:
Received 28 August 2016 5-Fluorouracil (5-FU) is a broad spectrum cytotoxic agent being used in chemotherapy of malignancies.
Received in revised form 28 October 2016 However, 5-FU shows a number of limitations like short half-life, non-selective biodistribution, and the
Accepted 3 November 2016 development of drug resistances by tumour cells. It was investigated the potential use of folic acid-
Available online 5 November 2016 decorated and PEGylated poly(D,L-lactide-co-glycolide) nanoparticles (FOL-PEG-PLGA NPs) for the
targeted delivery of 5-FU to colon and breast cancers. PEG-PLGA and FOL-PEG-PLGA conjugates were
Keywords: synthesized and characterized. NPs of PLGA, PEG-PLGA, and FOL-PEG-PLGA were prepared by
Cancer chemotherapy nanoprecipitation under optimal formulation conditions. They were found to be haemocompatible,
5-Fluorouracil
and exhibited negligible cytotoxicity in normal (CCD-18 and MCF-10A) and tumour (HT-29 and MCF-7)
FOL-PEG-PLGA conjugate
human cell lines. 5-FU loading capabilities were also dened, and the NPs exhibited an initial burst drug
Haemocompatibility
Ligand-receptor mediated drug delivery release followed by a sustained 5-FU release. In vitro cytotoxicity studies in folate-overexpressed HT-29
Nanoprecipitation. colon cancer cells and MCF-7 breast cancer cells demonstrated that the half maximal inhibitory
concentration (IC50) of 5-FU-loaded FOL-PEG-PLGA NPs was approximately 4-fold less than that of the 5-
FU-loaded PLGA NPs (p < 0.05). Consequently, FOL-PEG-PLGA NPs could have great potential as a targeted
5-FU delivery system for colon and breast cancer treatment.
2016 Elsevier B.V. All rights reserved.

1. Introduction Current therapeutic approaches in the management of cancer


include chemotherapy, surgery, radiation, and biological therapies,
Cancer is one of the leading causes of morbidity and mortality i.e. immune therapy and hormonal therapy. 5-uorouracil (5-FU), a
worldwide, with about 14 million new cases and 8.2 million deaths pyrimidine analogue, is among rst line cytotoxic agents employed
in 2012 (Stewart and Wild, 2014). According to the World Health in the chemotherapy of malignancies. 5-FU is a broad spectrum
Organization (WHO), the number of new cases is expected to molecule that interferes with nucleoside metabolism and can be
increase by around 70% rising from 14 million in 2012 to 22 million incorporated into ribonucleic acid (RNA) and deoxyribonucleic
within the next two decades. Among the most common causes of acid (DNA), mainly acting as a thymidylate synthase inhibitor and
cancer death, colorectal cancer (fourth) and breast cancer (fth) ultimately activating apoptosis in cancer cells (Longley et al.,
accounted for 694,000 and 521,000 deaths by year, respectively, in 2003). However, 5-FU shows a number of limitations including: i)
2012 (Stewart and Wild, 2014). short in vivo half-life due to rapid metabolism, thus sustained
therapeutic effects cannot be obtained; ii) erratic oral bioavailabil-
ity owing to metabolism by dihydropyrimidine dehydrogenase; iii)
wide non-selective in vivo distribution leading to toxic effects, e.g.
myelosupression and gastrointestinal, haematological, neural and
* Corresponding author at: Departamento de Farmacia y Tecnologa Farm- dermatological adverse effects; and, iv) the development of drug
acutica, Facultad de Farmacia, Universidad de Granada, 18071, Granada, Spain.
E-mail address: jlarias@ugr.es (J.L. Arias).
resistances by malignant cells.

http://dx.doi.org/10.1016/j.ijpharm.2016.11.012
0378-5173/ 2016 Elsevier B.V. All rights reserved.
62 M.M. El-Hammadi et al. / International Journal of Pharmaceutics 516 (2017) 6170

To address the above listed limitations, research has been 2. Materials and methods
focused on developing targeted delivery systems for 5-FU using
polymeric nanoparticles (NPs), such as poly(D,L-lactide-co-glyco- 2.1. Materials
lide) (PLGA) NPs. PLGA, a Food and Drug Administration (FDA)-
approved polymer, has shown very promising applications in drug PLGA (50:50; molecular weight average, Mw: 15,300 Da),
delivery given its biocompatibility, biodegradability, and versatili- hydroxyl-PEG-amine (Mw: 3000 Da), PEG bis(amine) (Mw:
ty (El-Hammadi and Arias, 2015; Holgado et al., 2011). Some 3000 Da), FOL, N,N0 -dicyclohexylcarbodiimide (DCC), N-hydroxy-
studies have reported the use of PLGA-based NPs to boost the succinimide (NHS), and triethylamine were purchased from
antitumour efciency of 5-FU against colon cancer (Shakeri-Zadeh Sigma-Aldrich (Munich, Germany). Anhydrous dichloromethane
et al., 2015; Tang et al., 2014; Wang et al., 2015) and breast cancer (DCM) and anhydrous dimethyl sulfoxide (DMSO) were purchased
(Balasubramanian et al., 2014; Nair et al., 2011; Zhu et al., 2016). from VWR (Barcelona, Spain). All other chemicals were of
While the properties of PLGA itself, such as the percentage of latic analytical quality (Panreac, Barcelona, Spain). Finally, deionized
acid and glycolic acid (Nair et al., 2011), can inuence the cytotoxic and ltered water was used in all the experiments (Milli-Q
effect of 5-FU, improved efciency and targeting characteristics Academic1, Millipore, Molsheim, France).
can be achieved by conjugation of PLGA with a hydrophilic polar
polymer, e.g. poly(ethylene glycol) (PEG) (Tang et al., 2014; Yadav 2.2. Methods
et al., 2010), and/or a targeting ligand, such as folic acid (FOL)
(Balasubramanian et al., 2014; Wang et al., 2015), hyaluronic acid 2.2.1. Synthesis of the FOL-PEG-PLGA conjugate
(Yadav et al., 2010), lactobionic acid (Dangi et al., 2014), The conjugate was synthesized in a 4-step process as previously
sulfadiazine (Guimares et al., 2015), transferrin (Balasubramanian described in the literature (Chen et al., 2012; Cheng et al., 2007;
et al., 2014), and translocator protein 18 kDa (Laquintana et al., Esmaeili et al., 2008; Saxena et al., 2012; Yoo and Park, 2004) with
2014). some modications.
PEG is a non-ionic hydrophilic polyether that it is extremely
biocompatible. Conjugation of PEG to PLGA-based NPs (the so- 2.2.1.1. Activation of PLGA. PLGA (1836 mg; 0.12 mmol), DCC
called PEGylation of the NPs) reports interesting advantages which (49.5 mg; 0.24 mmol) and NHS (27.6 mg; 0.24 mmol) were
include enhanced aqueous solubility, suppressed opsonization, dissolved in 4 mL of anhydrous DCM and left to stir at room
reduced aggregation and improved stability, minimized immuno- temperature under argon atmosphere for 24 h (PLGA/NHS/DCC
genicity, and prolonged biological half-life. For example, PEG-PLGA stoichiometric molar ratio: 1/2/2). The resultant solution was
NPs loaded with 5-FU have been shown to maintain drug release ltered, to remove the by-product dicyclohexylurea, and
over 5 days and to inhibit the peritoneal dissemination of colon precipitated by dropping into ice-cold ether. The solid
cancer in mice (Tang et al., 2014). precipitate was repeatedly washed in an ice-cold mixture of
FOL is a water-soluble vitamin B that it is essential for the ethyl ether and methanol to remove residual NHS. Ether was
production of new cells. A link has been established between low- decanted and the activated PLGA was completely dried under
folate diet and increased risk of a number of malignancies, vacuum.
including colon cancer (Fife et al., 2011) and breast cancer
(Manshadi et al., 2014). Folate receptors are overexpressed in 2.2.1.2. PEGylation of PLGA. The activated PLGA (385 mg;
some tumour cells, such as in colorectal carcinomas (D'Angelica 0.025 mmol) dissolved in 2 mL of anhydrous DCM was added
et al., 2011) and breast cancer (Zhang et al., 2014), and have a high drop-wise to PEG bis(amine) (225 mg; 0.075 mmol) dissolved in
afnity for FOL. Importantly, this afnity is maintained when FOL is 0.5 mL of anhydrous DCM. PEG bis(amine) was used in excess
bound with other molecules which enables targeting of folate- [PLGA/PEG bis(amine) stoichiometric molar ratio: 1/3] to suppress
linked drugs or NPs to folate receptor-expressing tumour cells. the formation of PLGA-PEG-PLGA triblock copolymers. The
A targeting delivery carrier, such as the one that combines PLGA reaction was carried out for 6 h with gentle stirring under argon
with PEG and FOL, is expected not to only optimize the anticancer atmosphere. The reaction mixture was then precipitated with cold
efciency of encapsulated 5-FU, but it can also overcome the drugs methanol and washed with the same solvent to remove unreacted
drawbacks through minimizing its toxicity, increasing its biological PEG. The precipitated product, amine-terminated diblock
(plasma) half-life, and overcoming the development of drug copolymer (PLGA-PEG-NH2), was ltered and dried under vacuum.
resistances. Our group has recently shown that the incorporation
of 5-FU in poly(e-caprolactone) NPs can considerably enhance the 2.2.1.3. Activation of FOL (formation of NHS-folate). FOL (220.8 mg;
activity of this drug against colon cancer cells and reduce their 0.5 mmol) was activated with DCC (206.4 mg; 1 mmol) and NHS
proliferation rate up to 40-times, as compared with the free (115 mg; 1 mmol) in 3 mL of DMSO in presence of 0.1 mL
anticancer drug (Ortiz et al., 2012). We have also developed 5-FU- triethylamine as a catalyst, under light protected argon
loaded magnetoliposomes for combined hyperthermia and che- atmosphere overnight. The solution was ltered to remove the
motherapy against colon cancer (Clares et al., 2013). dicyclohexylurea by-product, and then precipitated in ice-cold
In this work, we sought to synthesize a FOL-PEG-PLGA anhydrous ether. The product was obtained in the dry state
conjugate and use it to fabricate targeted NPs loaded with 5-FU following several steps of ether washing, decantation, and vacuum
for the treatment of colon and breast malignancies. 5-FU-loaded drying.
PLGAS PEG-PLGA, and FOL-PEG-PLGA NPs, prepared by nano-
precipitation, were completely characterized and studied. The 2.2.1.4. Conjugation of FOL to PEG-PLGA. Activated FOL (15.9 mg;
PLGA-based NPs were examined for their haemocompatibility as 0.036 mmol) and PEG-PLGA (329.4 mg; 0.018 mmol) were co-
well as cytotoxicity against colon and breast cancer cells. Despite dissolved in 3 mL of DMSO in light-protected conditions under
FOL-PEG-PLGA nanostructures have been postulated for controlled argon atmosphere at room temperature for 8 h. Excess amount of
and targeted delivery of drug molecules to malignant cells, i.e. activated FOL was used to get higher conjugation of FOL to PEG-
capecitabine (Wei et al., 2014), paclitaxel (Liang et al., 2011), PLGA. The reaction mixture was then mixed with 100 mL cold
saquinavir (Singh et al., 2015), to the best of our knowledge this is methanol and ltered through the paper lter. The precipitate on
the rst time that 5-FU-loaded FOL-PEG-PLGA NPs have shown the lter was dried under vacuum, and then dissolved in 50 mL
promising activity against colon and breast cancers. DCM; in this way, free FOL was precipitated in DCM but conjugated
M.M. El-Hammadi et al. / International Journal of Pharmaceutics 516 (2017) 6170 63

FOL was dissolved, after centrifugation at 21,000g for 30 min v) were placed on copper grids with formvar lm, and the grids
(Centrikon T-124 high-speed centrifuge; Kontron, Paris, France); were then dried at 25.0  0.5  C in a convection oven (J.P. Selecta, S.
the supernatant was dried under vacuum (Esmaeili et al., 2008). A., Abrera, Barcelona, Spain).
Folate content in the FOL-PEG-PLGA conjugate was computed Drug loading capacity was evaluated by determining the 5-FU
by dissolving a pre-weighed amount of FOL-PEG-PLGA in DMSO remained in the aqueous solution, collected after centrifugation of
and measuring the absorbance at 364 nm (UVvis Dinko spectro- the NPs dispersion, and deducting it from the total amount of the
photometer, Dinko, Barcelona, Spain). A calibration curve was used drug. Measurements were performed in triplicate using a
constructed using serially diluted concentrations of free FOL in previously validated ultraviolet-visible (UVvis) spectrophotomet-
DMSO (Yoo and Park, 2004). ric method at 266 nm (Ortiz et al., 2012). The incorporation of the
anticancer agent into the NPs was expressed in terms of drug
2.2.2. Synthesis of the PEG-PLGA conjugate entrapment efciency (EE, %) [(amount of 5-FU loaded [mg]/initial
The PEG-PLGA conjugate was synthesized following the rst 2 amount of 5-FU being used [mg])  100], and drug loading (DL, %)
steps of FOL-PEG-PLGA synthesis (see Section 2.2.1.). Briey, [(amount of 5-FU loaded [mg]/amount of drug-loaded NPs being
activated PLGA (704 mg; 0.046 mmol) dissolved in DCM was added obtained [mg])  100)].
drop-wise to a hydroxyl-PEG-amine (138 mg; 0.046 mmol) solu-
tion in DCM. The reaction was continued for 6 h and the product 2.2.5. Drug release studies
was precipitated with cold methanol, washed with methanol, Drug release experiments using formulations prepared under
ltered, and dried. optimal conditions (5-FU: 15 mg; PLGA: 50 mg; PVA: 1%; solvent
The synthesis of the new conjugates, i.e. PEG-PLGA and FOL- ratio: 5:10; sonication time: 0.5 min; sonication output: 20%; see
PEG-PLGA, was conrmed by proton nuclear magnetic resonance Sections 3.2. and 3.3.) were performed in phosphate buffered
(1H NMR) analysis (Bruker AM-300, Billerica, Massachusetts, USA), saline (PBS, pH 7.4  0.1) using the dialysis bag method. First, the
using deuterated chloroform (CDCl3) as solvent. dialysis membrane tubing (cut-off: 2 kDa; Spectrum1 Spectra/
Por1 6 dialysis membrane tubing, New Brunswick, New Jersey,
2.2.3. Preparation of the PLGA-based nanoparticles USA) was soaked in water at 25.0  0.5  C for 12 h prior to use. NPs
The NPs were fabricated by nanoprecipitation solvent evapora- dispersed in 2 mL of PBS (containing 5-FU at 0.5 mg/mL) were
tion technique using a probe sonicator (Branson Sonier 450, transferred into a dialysis bag and the two ends were fastened with
Emerson Electric Co., Danbury, Connecticut, USA). An organic clamps. The bag was placed in a beaker containing 200 mL of the
solution containing PLGA (50 mg; 3.268 mmol) (or an equivalent release medium, maintained at 37.0  0.5  C, and agitated at
amount of PEG-PLGA or FOL-PEG-PLGA, i.e. 59.8 mg or 60.4 mg, 200 rpm throughout the experiment. At selected time intervals
respectively) in acetone was added at once to 10 mL of an aqueous (0.25, 0.5, 1, 2, 3, 6, 9, 12, 24, 48, 72, 96, 120, and 144 h), 1 mL of the
solution of polyvinyl alcohol (PVA), and sonication was started medium was sampled and assayed at 266 nm 1 mL of PBS,
immediately. Prior to addition, the microtip of the sonicator was maintained at the same temperature, was added after each
immersed in the PVA solution, leaving 1 cm distance from the sampling to maintain the volume of the release medium and
bottom of the vial without touching its walls. An aluminum foil ensure sink conditions. The results were expressed as a drug
was placed over the opening of the vial to reduce the evaporative released percentage [(amount of 5-FU released in the medium
loss of the liquid. Pulsed mode, with a cycle of 50%, was employed [mg]/amount of 5-FU loaded in the NPs [mg])  100]. The
because it helps to avoid foaming, retards the rate of temperature experiments were performed in triplicate.
increase in the medium, and enables better temperature control
than continuous mode operation. For further temperature control, 2.2.6. In vitro blood compatibility study
the sample vial was surrounded with ice during the sonication The interaction of the blank (drug unloaded) NPs (PLGA NPs,
process. 5-FU-loaded NPs were prepared by dissolving the PEG-PLGA NPs, and FOL-PEG-PLGA NPs) with blood components
antitumour drug, at a given amount, in the organic phase prior was investigated to hypothesize their potential use for therapeutic
to addition to the aqueous phase. Finally, acetone was evaporated purposes, i.e. drug delivery against cancer. To that aim, blood
using a Bchi Rotavapor1 rotary evaporator (Bchi, Flawil, samples were obtained from three healthy female adults (24 to 41
Switzerland) to obtain an aqueous suspension of pure NPs. The years old), and poured into asks containing either ethylenedia-
dispersion was then centrifuged for 60 min at 10,000 rpm minetetraacetic acid (EDTA, used in the haemolysis and platelet
(Centrikon T-124 high-speed centrifuge; Kontron, Paris, France), activation experiments), or sodium citrate (used in the comple-
the supernatant removed, and the NPs re-suspended in water. ment system activation and plasma clotting time experiments).
To select the optimal formulation, several PVA concentrations Following a previously established procedure (Arias et al., 2011;
(0.5, 1, or 1.5%, w/v) and solvent ratios (3, 5, or 7 mL of the acetone Dash et al., 2010; Muoz de Escalona et al., 2016), the colloids were
organic solution) were tested. Sonication conditions were also placed in contact with blood aliquots to evaluate their effect on
studied by changing sonication time (0.5, 1, or 2 min) and sonicator erythrocytes, coagulation, and complement system. All the
output (20, 40, or 50%). The criteria for evaluating the produced determinations were performed in triplicate, and PBS was used
NPs were the particle size and the polydispersity index (PdI). as a negative control. The UVvis spectrophotometric methodolo-
gies were successfully validated and veried for accuracy,
2.2.4. Characterization of the PLGA-based nanoparticles precision, and linearity.
Mean particle size, size distribution (PdI), and zeta potential (z)
of the nanoformulations were determined using a Malvern 2.2.7. In vitro proliferation studies
Zetasizer (Nano-ZS, Malvern Instruments Ltd., Worcestershire, Normal human cell lines CCD-18 (colon broblast cells) and
UK). Cell temperature was 25.0  0.5  C with a detection angle of MCF-10A (breast epithelial cells) were obtained from the Scientic
90 . All measurements were performed in triplicate after Instrumentation Centre (University of Granada, Granada, Spain)
appropriate dilution of the NP dispersions in water ( 0.1%, w/v). and the American Type Culture Collection (ATCC, Manassas,
High resolution transmission electron microscopy (HRTEM; Virginia, USA), respectively. Human tumour cell lines HT-29
STEM CM20 microscope, Philips, Amsterdam, The Netherlands) (colonic adenocarcinoma cells) and MCF-7 (breast adenocarcino-
was employed to elucidate the shape, as well as to conrm the size ma cells) were obtained from ATCC (USA).
determinations of the NPs. Drops of dilute suspensions ( 0.1%, w/
64 M.M. El-Hammadi et al. / International Journal of Pharmaceutics 516 (2017) 6170

These cell lines were grown in a Dulbeccos modied Eagles The relative cell viability (%) was calculated as [(OD treated cells/
medium (DMEM, Sigma-Aldrich Co., Saint Louis, Missouri, USA) OD control (untreated) cells)  100].
supplemented with 10% heat-inactivated fetal bovine serum (FBS, Similarly, the MTT assay was further used to dene the
Lonza Group AG, Basel, Switzerland), 14 mM sodium bicarbonate, cytotoxicity of the formulations involving the drug, i.e. free 5-
15 mM 4-(2-hydroxyethyl)-1-piperazineethanesulfonic acid) FU, 5-FU-loaded PLGA NPs, 5-FU-loaded PEG-PLGA NPs, and 5-FU-
(HEPES), 2 mM L-glutamine, 40 mg/mL gentamicin, and 500 mg/ loaded FOL-PEG-PLGA NPs, against HT-29 and MCF-7 cells. The
mL ampicillin (Antibiticos de Len S.L.U., Len, Spain). They were concentration of these preparations in contact with cells ranged
seeded in monolayer culture into 96-well plates (6  103 cells/ from 0.1 to 50 mM equivalent 5-FU concentration.
well), and maintained until use at 37.0  0.5  C in a humidied 5%
CO2 incubator. 2.2.8. Statistical analysis
Cytotoxicity of blank (5-FU unloaded) PLGA-based NPs, i.e. The IBM SPSS Statistics software package (version 17; IBM
PLGA NPs, PEG-PLGA NPs, and FOL-PEG-PLGA NPs, was investigat- Corporation, Chicago, Illinois, USA) was used to perform all
ed not only in normal human cells (CCD-18 and MCF-10A) but also statistical analyses. Students t-test helped to compare results.
in the human tumour cell lines (HT-29 and MCF-7). To that aim, Data were expressed as means  standard deviations. Differences
quadruplicate cultures were evaluated by the 3-(4,5-dimethylth- were considered statistically signicant at a p value < 0.05.
iazol-2-yl)-3,5-diphenyl tetrazolium bromide (MTT) proliferation
assay, determining mitochondrial dehydrogenase activity. NP 3. Results and discussion
concentrations ranging from 0.1 to 200 mM (dilutions similar to
that of the drug-loaded particles used in these tests) were added to FOL-PEG-PLGA NPs were synthesized and investigated as a
the cells in the culture medium. After 72 h of incubation at potential carrier for 5-FU in the treatment of colon and breast
37.0  0.5  C, 20 mL of MTT solution were added to each well (5 mg/ cancers.
mL in cell culture medium). The culture medium was removed
after incubation during 4 h at this temperature, and the resultant 3.1. Characterization of the synthesized PEG-PLGA and FOL-PEG-PLGA
formazan crystals were dissolved in 200 mL of DMSO. Finally, the conjugates
optical density (OD) of converted dye, which is proportional to the
number of viable cells, was measured at 570 and 690 nm (Titertek PEG-PLGA and FOL-PEG-PLGA were synthesized by following a
multiscan colorimeter, Flow Laboratories, Irvine, California, USA). procedure previously described (Chen et al., 2012; Cheng et al.,

Fig. 1. Synthesis procedures followed to prepare the PEG-PLGA and FOL-PEG-PLGA conjugates.
M.M. El-Hammadi et al. / International Journal of Pharmaceutics 516 (2017) 6170 65

2007; Esmaeili et al., 2008; Saxena et al., 2012; Yoo and Park, 2004) does not compromise the ability of FOL to bind to folate receptor
with some modications (Fig. 1). The synthesis of the two (Sudimack and Lee, 2000). The folate terminal moiety has been
compounds involves the formation of amide bonds. These bonds found to reside on the surface when the NPs are formed (Low and
can be formed by a well-established and a relatively simple Kularatne, 2009).
chemical reaction. A succinimide end group is rst produced from Using 1H NMR analysis it was possible to identify the PLGA
a carboxyl group (COOH) in the presence of EDC, e.g. PLGA-COOH is moiety [-CH2- and CH3 peaks of poly(D,L-lactide), d = 5.2-5.3 and
converted to PLGA-NHS. Next, the succinimide end group is paired 1.5 ppm; CH2- peak of poly(glycolide), d = 4.8-4.9 ppm], the PEG
with an amine group (NH2). Upon conjugation of FOL to the moiety (d = 3.6 ppm), and the characteristic peaks of the folate
polymer, the succinimide derivative is obtained by conversion of moiety (d = 4.5, 6.6, 7.6, and 8.6 ppm) in the FOL-PEG-PLGA
the a-COOH group of FOL to the more reactive NHS group, which is conjugate (Fig. 2).
then reacted with the free primary NH2 group of the PEG-PLGA The conjugation percentage of FOL to PEG-PLGA was 42.3%
block copolymer. It is noteworthy that while a-COOH group (molar ratio) measured by UV absorbance determinations at
enables the preparation of the targeted conjugate, this conjugation 364 nm.

Fig. 2. 1H NMR spectra of: (a) PEG-PLGA conjugate, and (b) FOL-PEG-PLGA conjugate.
66 M.M. El-Hammadi et al. / International Journal of Pharmaceutics 516 (2017) 6170

Table 1
Effects of formulation, sonication, and drug loading on the characteristics of the PLGA NPs. The values are the mean  standard deviation (SD) of triplicate experiments.

Factors Formulation Sonication Drug used Size (nm) PdI z (mV)


studied (mg)
PVA concentration (%, Solvent ratio (organic to aqueous phase ratio, Time Output
w/v) o:w) (min) (%)
Formulation 0.5 3:10 0.5 20 0 207.3  6.8 0.063  0.011 23.5  3.1
5:10 210.8  19.1 0.081  0.035 27.3  2.3
7:10 230.1  26.8 0.291  0.016 26.1  1.7
1 3:10 184.2  15.4 0.044  0.022 24.9  2.1
5:10 181.3  11.7 0.058  0.018 25.5  1.9
7:10 210.1  24.2 0.202  0.033 23.5  2.3
1.5 3:10 180.6  11.4 0.051  0.015 27.3  2.6
5:10 181.8  12.9 0.671  0.032 26.1  2.2
7:10 195.9  20.2 0.166  0.034 24.1  1.7
Sonication 1 5:10 0.5 40 0 180.3  8.4 0.081  0.024 26.2  2.1
50 211.1  10.4 0.086  0.064 27.9  2.4
1 20 183.2  9.1 0.053  0.021 26.3  1.8
40 188.5  10.4 0.075  0.042 28.9  2.8
50 217.4  12.3 0.092  0.079 23.1  1.9
5 20 197.5  14.2 0.036  0.016 24.5  1.3
40 210.9  16.8 0.112  0.021 27.9  2.6
50 239.5  17.1 0.244  0.134 26.2  1.5
5-FU loading 1 5:1 0.5 20 1 181.4  10.3 0.049  0.022 24.1  2.9
3 184.9  7.2 0.059  0.023 27.7  1.9
5 185.8  11.9 0.056  0.026 24.4  2.9
8 187.6  7.1 0.062  0.011 28.3  2.1
10 186.2  10.3 0.064  0.015 23.6  1.4
12 188.4  9.3 0.071  0.013 26.9  2.6
15 190.7  13.1 0.063  0.019 25.1  2.6

3.2. Characterization of the PLGA-based nanoparticles higher percentage of 1.5% did not appear to add an additional effect
compared with the 1% PVA formulations. We also studied the effect
The NPs were produced successfully by dissolving PLGA, PEG- of altering the organic to aqueous phase ratio used. While particle
PLGA, or FOL-PEG-PLGA in acetone followed by nanoprecipitation size showed a trend to grow with the increase of solvent/water
in water. Characterization of the NPs was performed using dynamic ratio, the effect of this ratio was more obvious on PdI. At acetone/
light scattering (DLS) for particle size and z determinations, and water ratio of 7:10, the signicant increase in PdI, between 3 and 6
HRTEM for visualization of the NPs. folds as compared with the lower ratios, indicated a wider size
Effects of formulation, sonication, and drug loading on the distribution. This effect may be due to a partial attenuation and
characteristics of the PLGA NPs (i.e. particle size, PdI, and z) were dissipation of the applied ultrasonic energy by the increased
investigated (Table 1). In these studies only the examined volume of the medium.
parameter was varied while all the other parameters were kept Interestingly, short sonication time and lower output favoured
xed. When selecting formulation components, acetone was used the production of NPs with a smaller size and lower PdI. For
as the organic phase because it is a good solvent for 5-FU. In instance, optimal particles with an average size of 181.3 nm and a
addition, PVA was chosen as the stabilizer since it is commonly PdI of 0.058 were obtained by sonication for 0.5 min at 20% output,
employed in the preparation of PLGA NPs with a uniform size whereas sonication for 5 min at 50% output signicantly increased
distribution (Van de Ven et al., 2012). the average size to 239.5 nm (about 30%) and the PdI to 0.244
Among formulation parameters, we rst investigated varying (about 400%). Although cavitational collapse produced during
the percentage of the stabilizer, i.e. PVA. Increasing PVA from 0.5% sonication leads to particle size reduction, sonication-induced
to 1% (w/v) seemed to reduce the particle size and PdI, whereas a aggregates can form under certain conditions (Taurozzi et al.,

Fig. 3. HRTEM images of PLGA NPs (a), PEG-PLGA NPs (b), and FOL-PEG-PLGA NPs (c). Bar lengths: 200 nm (a), and 150 nm (b and c).
M.M. El-Hammadi et al. / International Journal of Pharmaceutics 516 (2017) 6170 67

2011). Increased collision frequency resulting from sonication and


augmented free energy owing to the increase in the liquid-solid
interface can enhance particleparticle interactions and favour
aggregate formation.
In general, drug loading into NPs is associated with an increase
in particle size. In this work, we observed a slight increase of up to
about 5% in the average particle size when increasing amounts of
5-FU, up to 15 mg, where used in NP preparation.
z values, ranging between 23 to 28 mV, did not seem to be
inuenced by formulation components, sonication conditions, nor
the amount of loaded drug.
Based on these ndings, the PVA concentration of 1%, the
organic to aqueous phase ratio of 5:10, sonication time of 0.5 min,
and sonication output of 20% were identied as parameters for the
optimal formulation of PLGA NPs. Under these parameters, the
mean particle size and PdI were found to be 181.3 nm and 0.058,
respectively. Furthermore, the volume of the organic solvent
(5 mL) was found to be sufcient to dissolve a suitable amount of 5-
FU.
The above dened optimal preparation conditions were also
used to obtain PEG-PLGA NPs and FOL-PEG-PLGA NPs character-
ized by a particle size of 145.3  10.4 nm and 149.2  7.0 nm,
respectively, with a PdI of 0.066  0.034 and 0.075  0.059,
correspondingly. These particle sizes were remarkably smaller
than those of the unmodied PLGA NPs. This result is in agreement
with previous ndings and it is thought to be due to the effect of
the hydrophilic PEG coating that improves the stability of the NPs
and results in a smaller particle size (Tao et al., 2012). Additionally,
while the negative surface charge of PEG-PLGA NPs, 22.3  2.1
mV, was similar to that of PLGA NPs (Table 1), FOL-PEG-PLGA NPs
had comparably a lower surface charge of 15.7  1.4 mV. This may
be owed to the effect of the protonated amino acid groups of folate
which diminish the negative charge of the PLGA carboxylic acid
groups (Liang et al., 2011).
HRTEM images conrmed the size determinations, and that
Fig. 4. Entrapment efciency (a) and loading (b) of 5-FU to the PLGA NPs (PLGA:
PLGA NPs, PEG-PLGA NPs, and FOL-PEG-PLGA NPs had similar 50 mg; PVA: 1%; solvent ratio: 5:10; sonication time: 0.5 min; sonication output:
morphologies (spherical shape, Fig. 3). 20%).

3.3. Drug loading studies

The EE (%) and DL (%) of 5-FU to the PLGA NPs as a function of


drug amount are collected in Fig. 4. As the drug amount used in NP
preparation was increased from 1 to 5 mg, the EE values increased
from 4.91  0.69 to 8.52  0.67 to reach a plateau at higher drug
amounts. Also, the loading percentage augmented linearly with
the amount of the drug and the maximum drug loading achieved
was 5.43  0.16 when 15 mg of drug were used. The relatively low
loading capacity may be due to the hydrophilic nature of 5-FU
which may encourage the immediate diffusion of the drug
molecules to the aqueous phase during preparation. In addition,
a relatively small amount of polymer (50 mg) was used in the
fabrication of the NPs which might also contribute to the resulting
loading capacity. Finally, similar drug incorporation results were
obtained when preparing 5-FU-loaded PEG-PLGA NPs and 5-FU-
loaded FOL-PEG-PLGA NPs. For instance, when 15 mg of 5-FU were
used, the 5-FU EE (%) values to the PEG-PLGA NPs and FOL-PEG-
PLGA NPs were 8.73  0.44 and 8.68  0.51, respectively, with a DL Fig. 5. In vitro release proles of 5-FU from: (^) PLGA NPs, (&) PEG-PLGA NPs, and
(%) of 5.52  0.14 and 5.49  0.15, correspondingly. (D) FOL-PEG-PLGA NPs, in PBS (pH 7.4  0.1) at 37.0  0.5  C. Data is presented as
mean value  standard deviation (SD) (n = 3).
3.4. In vitro drug release
approximately 2025% of the drug released after 1 h, followed
The in vitro release of 5-FU from the NPs was characterized in by a sustained, linear 5-FU release (Fig. 5). The initial burst release
PBS (pH 7.4  0.1) at 37.0  0.5  C, using a membrane dialysis of 5-FU could be due to the release of some weakly bound drug on
method. 5-FU-loaded PLGA NPs, 5-FU-loaded PEG-PLGA NPs, and the surface of the NPs which is rapidly released by diffusion.
5-FU-loaded FOL-PEG-PLGA NPs exhibited a similar drug release Whereas, the remained drug which is embedded in the NP
prole; characterized by an initial burst release, with structure was released slowly, likely due to polymer degradation
68 M.M. El-Hammadi et al. / International Journal of Pharmaceutics 516 (2017) 6170

by hydrolysis, in a more controlled manner over several days


(about 7580% was released over the 6-day experiment). Similar
release prole has been observed with PLGA NPs loaded with
different drugs (Menon et al., 2012; Park et al., 2009; Tewes et al.,
2007). However, PEG-PLGA NPs and FOL-PEG-PLGA NPs showed a
relatively higher 5-FU release compared with PLGA NPs; this
increased release was statically signicant (p value < 0.05) starting
from t = 2 h until the end of the experiment. The increase in drug
release may be attributed to variations in particle size, z values as
well as the greater hydrophilic surface of both PEG-PLGA NPs and
FOL-PEG-PLGA brought about by the PEG shell. The hydrophilic
surface can increase interaction with the aqueous medium, absorb
more water, and may subsequently induce faster degradation of
the NPs by hydrolysis which may lead to a faster 5-FU release. As a
result, 5-FU release from PEG-PLGA NPs and FOL-PEG-PLGA NPs
was completed in 4 days, while 6 days were needed by the PLGA
NPs to end the release of the loaded drug.

3.5. Haemocompatibility of the nanoparticles

Taking into consideration data in Table 2, the PLGA-based NPs


are expected to be characterized by a wide in vivo safety margin,
and suitability for parenteral administration (Arias et al., 2011;
Dash et al., 2010; Muoz de Escalona et al., 2016). In fact, a
negligible effect on haemolysis (even after 24 h), platelet activa-
tion, complement system activation, and plasma clotting time of
blood samples was observed (n = 3).

3.6. In vitro cytotoxicity experiments

As demonstrated in Fig. 6, blank (drug-unloaded) PLGA-based


NPs exhibited negligible cytotoxicity in normal (CCD-18, and MCF-
10A) and tumour (HT-29 and MCF-7) human cell lines at the
dilutions corresponding to that of 5-FU-loaded particles. Based on
these ndings, and also considering the data coming from blood
compatibility tests (see Section 3.5.), it can be assumed that these
polymeric nanosystems present an adequate biocompatibility and
safety for drug delivery purposes.
Fig. 7 displays the in vitro cytotoxic effect of free 5-FU, and 5-
FU-loaded NPs in HT-29 and MCF-7 cells. These formulations Fig. 6. In vitro cytotoxicity of PLGA NPs (a), PEG-PLGA NPs (b), and FOL-PEG-PLGA
inhibited cell proliferation in a dose-dependent manner. In both NPs (c) in CCD-18 human colon broblast cells, HT-29 human colon cancer cells,
MCF-10A human breast epithelial cells, and MCF-7 human breast cancer cells, after
cell lines, the half maximal inhibitory concentration (IC50) values of
72 h of exposure to a wide range of NP concentrations: 0.1 mM (grey column), 50 mM
5-FU-loaded PEG-PLGA NPs and 5-FU-loaded FOL-PEG-PLGA NPs (light grey column), 100 mM (black column), and 200 mM (white column). The
were approximately 2-fold and 4-fold, respectively, less than that values are the mean  standard deviation (SD) of quadruplicate cultures. Cells
of the 5-FU-loaded PLGA NPs (p < 0.05), whereas the IC50 of 5-FU- without treatment were used as control to calculate the relative cell viability (%).
loaded PLGA NPs was found to be similar to that of the free
antitumour agent (Fig. 7). The lower cytotoxicity of free 5-FU may (Pamujula et al., 2012). This effect was associated with the smaller
be attributed to the drug resistance resulting in the efux of 5-FU particle size of the PEGylated PLGA NPs, and similar ndings have
molecules by P-glycoprotein (P-gp) pumps (Fojo and Coley, 2007). been described in Section 3.2. on the effect of PEGylation on the
In addition, the higher cytotoxicity of 5-FU-loaded PEG-PLGA NPs size of the PLGA particles. The positive effect of PEGylation on NP
compared to 5-FU-loaded PLGA NPs is in agreement with a recent uptake by cancer cells overexpressing P-gp has been further
study in which PEGylation of PLGA NPs was found to signicantly described in liposomes (Kopecka et al., 2014) and poly(amido
increase cellular uptake by 4T1 murine breast cancer cells amine) (PAMAM) dendrimers (Liu and Chiu, 2013).

Table 2
Blood compatibility of blank (5-FU unloaded) PLGA-based NPs in terms of haemolysis (%), platelet activation (soluble P-selectin release, ng/mL), complement system
activation (C3a release: C3a desArg, ng/mL), and plasma recalcication time (T1/2max, min). Data is indicated as mean value  standard deviation (SD) (n = 4).

Sample Haemolysis (%) soluble P-selectin release (ng/mL) C3a desArg (ng/mL) T1/2max (min)

Incubation time

2h 4h 8h 24 h
PLGA NPs 0.8  0.1 1.1  0.2 1.9  0.2 1.8  0.4 103  4 296  13 12.8  1.6
PEG-PLGA NPs 1.1  0.3 1.2  0.4 0.9  0.3 1.1  0.1 105  3 301  14 11.3  1.4
FOL-PEG-PLGA NPs 1.2  0.2 0.9  0.3 1.7  0.4 1.9  0.3 101  5 299  11 13.1  1.5
Control (PBS solution) 0 0 0 0 98  7 290  11 10.9  2.1
M.M. El-Hammadi et al. / International Journal of Pharmaceutics 516 (2017) 6170 69

the FOL-decorated and PEGylated PLGA NPs may facilitate targeted


delivery of 5-FU to colon and breast tumours.

Acknowledgements and funding

Financial support of the Erasmus Mundus JOSYLEEM Program


is acknowledged as is the post-doctoral fellowship to Mazen M. El-
Hammadi. The authors gratefully acknowledge the assistance of Dr.
Ana Conejo Garca (Department of Organic Chemistry, Faculty of
Pharmacy, University of Granada, Spain) in the chemical syntheses
carried out in this work. Research leading to these results has
received funding from FEDER, Plan Nacional de Investigacin
Cientca, Desarrollo e Innovacin Tecnolgica (I + D + i), Instituto
de Salud Carlos III (FIS, Spain): projects PI11/01862 and PI11/02571;
and from Junta de Andaluca (Spain, project PE-2012-FQM-694).

References

Arias, J.L., Lpez-Viota, M., Sez-Fernndez, E., Ruiz, M.A., Delgado, A.V., 2011.
Engineering of an antitumor (core/shell) magnetic nanoformulation based on
the chemotherapy agent ftorafur. Colloids Surf. A Physicochem. Eng. Aspects.
384, 157163.
Balasubramanian, S., Girija, A.R., Nagaoka, Y., Iwai, S., Suzuki, M., Kizhikkilot, V.,
Yoshida, Y., Maekawa, T., Nair, S.D., 2014. Curcumin and 5-uorouracil-loaded,
folate- and transferrin-decorated polymeric magnetic nanoformulation: a
synergistic cancer therapeutic approach, accelerated by magnetic
hyperthermia. Int. J. Nanomed. 9, 437459.
Chen, J., Li, S., Shen, Q., 2012. Folic acid and cell-penetrating peptide conjugated
PLGA-PEG bifunctional nanoparticles for vincristine sulfate delivery. Eur. J.
Pharm. Sci. 47, 430443.
Cheng, J., Teply, B.A., Sheri, I., Sung, J., Luther, G., Gu, F.X., Levy-Nissenbaum, E.,
Radovic-Moreno, A.F., Langer, R., Farokhzad, O.C., 2007. Formulation of
functionalized PLGA-PEG nanoparticles for in vivo targeted drug delivery.
Fig. 7. In vitro cytotoxicity of free 5-FU (grey column), 5-FU-loaded PLGA NPs (light Biomaterials 28, 869876.
grey column), 5-FU-loaded PEG-PLGA NPs (black column), and 5-FU-loaded FOL- Clares, B., Biedma-Ortiz, R.A., Sez-Fernndez, E., Prados, J.C., Melguizo, C., Cabeza,
L., Ortiz, R., Arias, J.L., 2013. Nano-engineering of 5-uorouracil-loaded
PEG-PLGA NPs (white column) in HT-29 human colon cancer cells (a) and (b) MCF-7
magnetoliposomes for combined hyperthermia and chemotherapy against
human breast cancer cells, after 72 h of exposure to a wide range of NP
colon cancer. Eur. J. Pharm. Biopharm. 85, 329338.
concentrations (up to 25 mM equivalent 5-FU concentration). The values are the D'Angelica, M., Ammori, J., Gonen, M., Klimstra, D.S., Low, P.S., Murphy, L., Weiser, M.
mean  standard deviation (SD) of quadruplicate cultures. Cells without treatment R., Paty, P.B., Fong, Y., Dematteo, R.P., Allen, P., Jarnagin, W.R., Shia, J., 2011. Folate
were used as control to calculate the relative cell viability (%). Insets: IC50 values receptor-alpha expression in resectable hepatic colorectal cancer metastases:
(mM) of the 5-FU-based formulations. patterns and signicance. Mod. Pathol. 24, 12211228.
Dangi, R., Hurkat, P., Jain, A., Shilpi, S., Jain, A., Gulbake, A., Jain, S.K., 2014. Targeting
liver cancer via ASGP receptor using 5-FU-loaded surface-modied PLGA
Finally, the cytotoxic effect of 5-FU was found to be additionally
nanoparticles. J. Microencapsul. 31, 479487.
increased with the FOL-decorated and PEGylated (FOL-PEG-PLGA) Dash, B.C., Rthor, G., Monaghan, M., Fitzgerald, K., Gallagher, W., Pandit, A., 2010.
NPs (Fig. 7). These results may be associated to an enhanced The inuence of size and charge of chitosan/polyglutamic acid hollow spheres
cellular uptake via folate receptor mediated endocytosis, thus on cellular internalization, viability and blood compatibility. Biomaterials 31
88188197.
avoiding 5-FU efux by P-gp and leading to a signicantly El-Hammadi, M., Arias, J.L., 2015. Advanced engineering approaches in the
augmented effect against the folate receptor-overexpressed HT- development of PLGA-based nanomedicines. In: Aliofkhazraei, M. (Ed.),
29 and MCF-7 cells, as previously reported in the literature Handbook of Nanoparticles. Springer International Publishing, Switzerland, pp.
10091039.
(Balasubramanian et al., 2014; Wang et al., 2015). The FOL-PEG- Esmaeili, F., Ghahremani, M.H., Ostad, S.N., Atyabi, F., Seyedabadi, M., Malekshahi,
PLGA nanosystem being described here has efcaciously improved M.R., Amini, M., Dinarvand, R., 2008. Folate-receptor-targeted delivery of
5-FU activity against MCF-7 cells as compared to alternative pure docetaxel nanoparticles prepared by PLGA-PEG-folate conjugate. J. Drug Target.
16, 415423.
PLGA NPs (Nair et al., 2011) and against HT-29 cells as compared to Fife, J., Raniga, S., Hider, P.N., Frizelle, F.A., 2011. Folic acid supplementation and
alternative pure FOL-decorated (non-PEGylated) PLGA NPs (Wang colorectal cancer risk: a meta-analysis. Colorectal Dis. 13, 132137.
et al., 2015). Fojo, T., Coley, H.M., 2007. The role of efux pumps in drug-resistant metastatic
breast cancer: new insights and treatment strategies. Clin. Breast Cancer 7, 749
756.
4. Conclusions Guimares, P.P., Oliveira, S.R., de Castro Rodrigues, G., Gontijo, S.M., Lula, I.S., Corts,
M.E., Denadai, .M., Sinisterra, R.D., 2015. Development of sulfadiazine-
decorated PLGA nanoparticles loaded with 5-uorouracil and cell viability.
It has been reported for the rst time the use of FOL-PEG-PLGA
Molecules 20, 879899.
NPs as a targeted delivery system of 5-FU against colon and breast Holgado, M.A., Alvarez-Fuentes, J., Fernndez-Arvalo, M., Arias, J.L., 2011.
cancers. To that aim, a PEG-PLGA copolymer was synthesized and Possibilities of poly(D,L-lactide-co-glycolide) in the formulation of
functionalized with FOL, which was then formulated as NPs by nanomedicines against cancer. Curr. Drug Targets 12, 10961111.
Kopecka, J., Salzano, G., Campia, I., Lusa, S., Ghigo, D., De Rosa, G., Riganti, C., 2014.
nanoprecipitation. 5-FU loading conditions resulted in an adequate Insights in the chemical components of liposomes responsible for P-
drug vehiculization by the NPs and in sustained (biphasic) drug glycoprotein inhibition. Nanomedicine 10, 7787.
release properties. FOL-PEG-PLGA NPs entrapping 5-FU showed a Laquintana, V., Denora, N., Lopalco, A., Lopedota, A., Cutrignelli, A., Lasorsa, F.M.,
Agostino, G., Franco, M., 2014. Translocator protein ligand-PLGA conjugated
superior cytotoxicity over non-targeted PLGA-based NPs in folate nanoparticles for 5-uorouracil delivery to glioma cancer cells. Mol. Pharm. 11,
receptor-overexpressed HT-29 colon and MCF-7 breast cancer cell 859871.
lines. On the other hand, blank (drug unloaded) NPs were Liang, C., Yang, Y., Ling, Y., Huang, Y., Li, T., Li, X., 2011. Improved therapeutic effect of
folate-decorated PLGA-PEG nanoparticles for endometrial carcinoma. Bioorg.
haemocompatible with negligible cytotoxicity, making them Med. Chem. 19, 40574066.
promising candidates as drug carriers. Our ndings imply that
70 M.M. El-Hammadi et al. / International Journal of Pharmaceutics 516 (2017) 6170

Liu, Y., Chiu, G.N., 2013. Dual-functionalized PAMAM dendrimers with improved P- World cancer report. In: Stewart, B.W., Wild, C.P. (Eds.), France: International
glycoprotein inhibition and tight junction modulating effect. Agency for Research on Cancer. World Health Organization.
Biomacromolecules 14, 42264235. Sudimack, J., Lee, R.J., 2000. Targeted drug delivery via the folate receptor. Adv. Drug
Longley, D.B., Harkin, D.P., Johnston, P.G., 2003. 5-uorouracil: mechanisms of Deliv. Rev. 41, 147162.
action and clinical strategies. Nat. Rev. Cancer 3, 330338. Tang, Q., Wang, Y., Huang, R., You, Q., Wang, G., Chen, Y., Jiang, Z., Liu, Z., Yu, L.,
Low, P.S., Kularatne, S.A., 2009. Folate-targeted therapeutic and imaging agents for Muhammad, S., Wang, X., 2014. Preparation of anti-tumor nanoparticle and its
cancer. Curr. Opin. Chem. Biol. 13, 256262. inhibition to peritoneal dissemination of colon cancer. PLoS One 9, e98455.
Manshadi, S., Ishiguro, L., Sohn, K., Medline, A., Renlund, R., Croxford, R., Kim, Y., Tao, Y., Han, J., Dou, H., 2012. Surface modication of paclitaxel-loaded polymeric
2014. Folic acid supplementation promotes mammary tumor progression in a nanoparticles: evaluation of in vitro cellular behavior and in vivo
rat model. PLoS One 9, e84635. pharmacokinetic. Polymer 53, 50785086.
Menon, J.U., Kona, S., Wadajkar, A.S., Desai, F., Vadla, A., Nguyen, K.T., 2012. Effects of Taurozzi, J.S., Hackley, V.A., Wiesner, M.R., 2011. Ultrasonic dispersion of
surfactants on the properties of PLGA nanoparticles. J. Biomed. Mater. Res. A nanoparticles for environmental, health and safety assessment-issues and
100, 19982005. recommendations. Nanotoxicology 5, 711729.
Muoz de Escalona, M., Sez-Fernndez, E., Prados, J.C., Melguizo, C., Arias, J.L., 2016. Tewes, F., Munnier, E., Antoon, B., Ngaboni Okassa, L., Cohen-Jonathan, S., Marchais,
Magnetic solid lipid nanoparticles in hyperthermia against colon cancer. Int. J. H., Douziech-Eyrolles, L., Souc, M., Dubois, P., Chourpa, I., 2007. Comparative
Pharm. 504, 1119. study of doxorubicin-loaded poly(lactide-co-glycolide) nanoparticles prepared
Nair, K.L., Jagadeeshan, S., Nair, S.A., Kumar, G.S., 2011. Biological evaluation of 5- by single and double emulsion methods. Eur. J. Pharm. Biopharm. 66, 488492.
uorouracil nanoparticles for cancer chemotherapy and its dependence on the Van de Ven, H., Paulussen, C., Feijens, P.B., Matheeussen, A., Rombaut, P., Kayaert, P.,
carrier, PLGA. Int. J. Nanomed. 6, 16851697. Van den Mooter, G., Weyenberg, W., Cos, P., Maes, L., Ludwig, A., 2012. PLGA
Ortiz, R., Prados, J., Melguizo, C., Arias, J.L., Ruiz, M.A., Alvarez, P.J., Caba, O., Luque, R., nanoparticles and nanosuspensions with amphotericin B: potent in vitro and in
Segura, A., Arnega, A., 2012. 5-Fluorouracil-loaded poly(e-caprolactone) vivo alternatives to Fungizone and AmBisome. J. Control. Release 161, 795803.
nanoparticles combined with phage E gene therapy as a new strategy against Wang, Y., Li, P., Chen, L., Gao, W., Zeng, F., Kong, L.X., 2015. Targeted delivery of 5-
colon cancer. Int. J. Nanomed. 7, 95107. uorouracil to HT-29 cells using high efcient folic acid-conjugated
Pamujula, S., Hazari, S., Bolden, G., Graves, R.A., Chinta, D.D., Dash, S., Kishore, V., nanoparticles. Drug Deliv. 22, 191198.
Mandal, T.K., 2012. Cellular delivery of PEGylated PLGA nanoparticles. J. Pharm. Wei, K., Peng, X., Zou, F., 2014. Folate-decorated PEG-PLGA nanoparticles with silica
Pharmacol. 64, 6167. shells for capecitabine controlled and targeted delivery. Int. J. Pharm. 464, 225
Park, J., Fong, P.M., Lu, J., Russell, K.S., Booth, C.J., Saltzman, W.M., Fahmy, T.M., 2009. 233.
PEGylated PLGA nanoparticles for the improved delivery of doxorubicin. Yadav, A.K., Agarwal, A., Rai, G., Mishra, P., Jain, S., Mishra, A.K., Agrawal, H., Agrawal,
Nanomedicine 5, 410418. G.P., 2010. Development and characterization of hyaluronic acid decorated PLGA
Saxena, V., Naguib, Y., Hussain, M.D., 2012. Folate receptor targeted 17-allylamino- nanoparticles for delivery of 5-uorouracil. Drug Deliv. 17, 561572.
17-demethoxygeldanamycin (17-AAG) loaded polymeric nanoparticles for Yoo, H.S., Park, T.G., 2004. Folate receptor targeted biodegradable polymeric
breast cancer. Colloids Surf. B Biointerfaces 94, 274280. doxorubicin micelles. J. Control. Release 96, 273283.
Shakeri-Zadeh, A., Khoee, S., Shiran, M.-B., Shari, A.M., Khoei, S., 2015. Synergistic Zhang, Z., Wang, J., Tacha, D.E., Li, P., Bremer, R.E., Chen, H., Wei, B., Xiao, X., Da, J.,
effects of magnetic drug targeting using a newly developed nanocapsule and Skinner, K., Hicks, D.G., Bu, H., Tang, P., 2014. Folate receptor a associated with
tumor irradiation by ultrasound on CT26 tumors in BALB/c mice. J. Mater. Chem. triple-negative breast cancer and poor prognosis. Arch. Pathol. Lab. Med. 138,
B 3, 18791887. 890895.
Singh, R., Kesharwani, P., Mehra, N.K., Singh, S., Banerjee, S., Jain, N.K., 2015. Zhu, W., Lee, S.J., Castro, N.J., Yan, D., Keidar, M., Zhang, L.G., 2016. Synergistic effect
Development and characterization of folate anchored Saquinavir entrapped of cold atmospheric plasma and drug loaded core-shell nanoparticles on
PLGA nanoparticles for anti-tumor activity. Drug Dev. Ind. Pharm. 41, 1888 inhibiting breast cancer cell growth. Sci. Rep. 6, 21974.
18901.

S-ar putea să vă placă și