ArticlePDF Available

A toxic organic solvent-free technology for the preparation of PEGylated paclitaxel nanosuspension based on human serum albumin for effective cancer therapy

Taylor & Francis
International Journal of Nanomedicine
Authors:

Abstract and Figures

Clinically, paclitaxel (PTX) is one of most commonly prescribed therapies against a wide range of solid neoplasms. Despite its success, the clinical applicability of PTX (Taxol®) is severely hampered by systemic toxicities induced by Cremophor EL. While attempts to bypass the need for Cremophor EL have been developed through platforms such as Abraxane™, nab™ relies heavily on the use of organic solvents, namely, chloroform. The toxicity introduced by residual chloroform poses a potential risk to patient health. To mitigate the toxicities of toxic organic solvent-based manufacture methods, we have designed a method for the formulation of PTX nanosuspensions (PTX-PEG [polyethylene glycol]-HSA [human serum albumin]) that eliminates the dependence on toxic organic solvents. Coined the solid-dispersion technology, this technique permits the dispersion of PTX into PEG skeleton without the use of organic solvents or Cremophor EL as a solubilizer. Once the PTX-PEG dispersion is complete, the dispersion can be formulated with HSA into nanosuspensions suitable for intravenous administration. Additionally, the incorporation of PEG permits the prolonged circulation through the steric stabilization effect. Finally, HSA-mediated targeting permits active receptor-mediated endocytosis for enhanced tumor uptake and reduced side effects. By eliminating the need for both Cremophor EL and organic solvents while simultaneously increasing antitumor efficacy, this method provides a superior alternative to currently accepted methods for PTX delivery.
This content is subject to copyright. Terms and conditions apply.
© 2015 Yin et al. This work is published by Dove Medical Press Limited, and licensed under Creative Commons Attribution – Non Commercial (unported, v3.0)
License. The full terms of the License are available at http://creativecommons.org/licenses/by-nc/3.0/. Non-commercial uses of the work are permitted without any further
permission from Dove Medical Press Limited, provided the work is properly attributed. Permissions beyond the scope of the License are administered by Dove Medical Press Limited. Information on
how to request permission may be found at: http://www.dovepress.com/permissions.php
International Journal of Nanomedicine 2015:10 7397–7412
International Journal of Nanomedicine Dovepress
submit your manuscript | www.dovepress.com
Dovepress 7397
ORIGINAL RESEARCH
open access to scientific and medical research
Open Access Full Text Article
http://dx.doi.org/10.2147/IJN.S92697
A toxic organic solvent-free technology for the
preparation of PEGylated paclitaxel nanosuspension
based on human serum albumin for effective cancer
therapy
Tingjie Yin*
Lihui Dong*
Bei Cui
Lei Wang
Lifang Yin
Jianping Zhou
Meirong Huo
Sta te Key L abo rat ory of Natural
Medicines, Department of
Pharmaceutics, China Pharmaceutical
University, Nanjing, People’s Republic
of China
*These authors contributed equally
to this work
Abstract: Clinically, paclitaxel (PTX) is one of most commonly prescribed therapies against a
wide range of solid neoplasms. Despite its success, the clinical applicability of PTX (Taxol®) is
severely hampered by systemic toxicities induced by Cremophor EL. While attempts to bypass
the need for Cremophor EL have been developed through platforms such as Abraxane™, nab™
relies heavily on the use of organic solvents, namely, chloroform. The toxicity introduced by
residual chloroform poses a potential risk to patient health. To mitigate the toxicities of toxic
organic solvent-based manufacture methods, we have designed a method for the formulation
of PTX nanosuspensions (PTX-PEG [polyethylene glycol]-HSA [human serum albumin]) that
eliminates the dependence on toxic organic solvents. Coined the solid-dispersion technology,
this technique permits the dispersion of PTX into PEG skeleton without the use of organic
solvents or Cremophor EL as a solubilizer. Once the PTX-PEG dispersion is complete, the
dispersion can be formulated with HSA into nanosuspensions suitable for intravenous admin-
istration. Additionally, the incorporation of PEG permits the prolonged circulation through the
steric stabilization effect. Finally, HSA-mediated targeting permits active receptor-mediated
endocytosis for enhanced tumor uptake and reduced side effects. By eliminating the need for
both Cremophor EL and organic solvents while simultaneously increasing antitumor efficacy,
this method provides a superior alternative to currently accepted methods for PTX delivery.
Keywords: human serum albumin, nanosuspension, paclitaxel, polyethylene glycol, solid-
dispersion technology
Introduction
Paclitaxel (PTX) is a clinically accepted front-line therapy against a wide range of
solid neoplasms since 1990. It works by promoting microtubule polymerization from
tubulin heterodimers, but inhibiting microtubule destruction. This leads to the induction
of cell apoptosis at the late G2/M phase.1 Despite its wide use, the practical applica-
tion of PTX is still limited by its poor aqueous solubility and low therapeutic index.2
In its commercial formulations (Taxol®, Bristol-Myers Squibb, New York, NY, USA),
PTX is formulated in a 50:50 mixture of Cremophor EL and dehydrated ethanol.3,4
Unfortunately, Cremophor EL has been attributed to significant side effects, such as
hypersensitivity, neurotoxicity, nephrotoxicity, and cardiotoxicity when intravenously
(IV) administrated at a large dose.5,6
Because of the inherent problems associated with Cremophor EL, a number of
alternative formulations including liposomes,7 lipid emulsions,8 polymeric micelles,9
Correspondence: Jianping Zhou;
Meirong Huo
State Key Laboratory of Natural
Medicines, Department of Pharmaceutics,
China Pharmaceutical University,
24 Tongjiaxiang, Nanjing 210009,
People’s Republic of China
Tel +86 25 8327 1102
Fax +86 25 8330 1606
Email cpu_zhoujp@163.com;
huomeirongcpu@163.com
Journal name: International Journal of Nanomedicine
Article Designation: Original Research
Year: 2015
Volume: 10
Running head verso: Yin et al
Running head recto: A toxic organic solvent-free technology for preparation of PTX-PEG-HSA
DOI: http://dx.doi.org/10.2147/IJN.S92697
Number of times this article has been viewed
This article was published in the following Dove Press journal:
International Journal of Nanomedicine
11 December 2015
International Journal of Nanomedicine 2015:10
submit your manuscript | www.dovepress.com
Dovepress
Dovepress
7398
Yin et al
cyclodextrin complexes,10 polymer–PTX conjugates,11
nanosuspensions,12 and nanoparticles (NPs)13 have been inves-
tigated to circumvent the need for Cremophor EL. Among
these alternatives, nanosuspensions in particular offer unique
advantages. Some examples include increased solubility for
hydrophobic drugs, greater bioavailability in vivo, improved
particle accumulation through the enhanced permeability and
retention (EPR) effect, excellent applicability to most hydro-
phobic drugs, and suitability for industrial scale-up.14 Conse-
quently, nanosuspension has been widely used as an effective
drug delivery system for hydrophobic anticancer drugs.
While nanosuspensions offer a solution for the chemical
formulation of hydrophobic drugs, targeting remains an issue
that still must be addressed. Human serum albumin (HSA)-
mediated targeting uses one of the major soluble proteins
in the blood circulation, HSA, and has been reported as a
promising biocompatible method for drug delivery.15 Biologi-
cally, HSA functions to maintain colloidal osmotic pressure
in the blood, alongside facilitating the transport of several
endogenous and exogenous substances.16,17 However, HSA
also naturally possesses specific targeting abilities to inflamed
and malignant tissues. Once targeted to the tumor site, HSA
is efficiently transported into tumor cells through gp60
(a 60 KDa glycoprotein) or SPARC (one secreted protein,
acidic and rich in cysteine) receptor-mediated endocytosis.18,19
As a result, HSA has been exploited in a multitude of drug
delivery systems due to its natural tumor targeting ability
and absence of toxicity or immunogenicity.19
Using nanosuspensions to mediate the chemical for-
mulation of PTX and HSA for tumor targeting, Ameri-
can Bioscience Inc. (Blauvelt, NY, USA) produced the
first biodegradable HSA-based PTX nanosuspension,
Abraxane™, by nab™. Abraxane™ is the first US Food and
Drug Administration (FDA)-approved chemotherapeutic
formulation based on nanotechnology.20 To date, the main
preparation methods of albumin-based formations can be
concluded into four techniques: emulsification,21 thermal
gelation,22 desolvation (coacervation),23 and nab™.24 While
the first three conventional techniques involve chemical
cross-linking processes or heat denaturation, nab™ oxidizes
sulfhydryl residues in albumin through homogenization to
form new cross-linking disulfide bonds, without denaturing
albumin. nab™ provides a unique window of opportunity for
albumin-based formulation, as it circumvents the destruction
of albumin’s biological characteristic while not requiring the
use of chemical crosslinkers. The lack of crosslinkers ensures
the absence of aldehyde byproducts, a prominent defect in
the conventional preparation methods.25,26
Albeit promising, the use of chloroform in nab™
manufacturing leads to chronic toxicities. In response to
the issue of chronic toxicities, we have developed a method
for the successful formulation of PTX nanosuspensions
that eliminates the dependence on toxic organic solvents
during manufacturing. Furthermore, NPs formed using this
method still retain their suitability for IV administration.
Polyethylene glycol (PEG) is a highly hydrophilic polymer
with superb biocompatibility and biodegradability, which has
been widely used as a hydrophilic solubilizing regent and
pharmacokinetic tailor.27,28 Therefore, PEG was employed to
effectively disperse PTX.29,30 In detail, PTX is first co-melted
with PEG to form a solid dispersion, which was then mixed
with the HSA solution and passed under a high pressure jet
to form the PTX-PEG-HSA nanosuspension with a narrow
size distribution. Because of the “steric stabilization” effect
of PEG, PTX-PEG-HSA nanosuspensions remain invisible
to the reticuloendothelial system (RES). As a result, PTX-
PEG-HSA nanosuspensions exhibit significantly prolonged
circulation and enhanced tumor accumulation while mitigat-
ing collateral damage to noncancerous tissues. To confirm
these advantages, the properties of the PTX-PEG-HSA nano-
suspension such as drug-loading (DL) capacity, pH, osmotic
pressure, compatible stability, drug release, and cell growth
inhibition against MDA-MB-231 cells were characterized
in vitro. Furthermore, the in vivo antitumor efficacy and
systemic toxicity of PTX-PEG-HSA were further evaluated
in MDA-MB-231 tumor-bearing nude mice in comparison
to PTX-HSA and Taxol®.
Materials and methods
Materials
HSA with a molecular weight (MW) of 65,000 Da was
purchased from Yumin Biotech Co., Ltd. (Shanghai,
People’s Republic of China). PTX and PEG (MW 6 kDa)
were purchased from Sunve Pharmaceutical Co., Ltd.
(Shanghai, People’s Republic of China) and Taihua Natural
Plant Pharmaceutical Co., Ltd. (Xi’an, Shanxi Province,
People’s Republic of China), respectively. Diazepam was
obtained from Nanjing Xiandao Chemical Co., Ltd. (Jiangsu,
People’s Republic of China). 3-(4,5-Dimethylthiazol-2-yl)-
2,5-diphenyltetrazolium bromide (MTT) was purchased
from Sigma-Aldrich (St Louis, MO, USA). The Cy5.5
N-hydroxysuccinimide ester (Cy5.5-NHS) and fluorescein
isothiocyanate (FITC) were obtained from Beijing Fanbo
Science and Technology Co., Ltd. (Beijing, People’s Republic
of China). HPLC-grade reagents were used as the mobile
phase in HPLC (high-performance liquid chromatography)
International Journal of Nanomedicine 2015:10 submit your manuscript | www.dovepress.com
Dovepress
Dovepress
7399
A toxic organic solvent-free technology for preparation of PTX-PEG-HSA
analysis, and all other reagents were of analytical grade and
used without further purification. Distilled and deionized
water was used in all experiments.
Cells
MDA-MB-231 cells were provided by Origin Biosciences
Inc. (Nanjing, People’s Republic of China). Cells were
cultured in Dulbecco’s Modified Eagle’s Medium supple-
mented with 10% fetal bovine serum at 37°C in a 5% CO2
atmosphere.
Animals
BALB/c nude mice were obtained from Shanghai Institute of
Materia Medica, Chinese Academy of Sciences, Shanghai,
People’s Republic of China. All the animals were pathogen-
free and allowed to access food and water freely. All animals
were treated in accordance with the Guide for Care and Use
of Laboratory Animals, approved by China Pharmaceutical
University.
Preparation of PTX-PEG-HSA and PTX-
HSA nanosuspension
Preparation of PTX-PEG-HSA nanosuspension
First, the solid dispersion technique was used for the prepara-
tion of PTX-PEG suspension. PTX (5 mg) was cofused with
PEG (10, 15, 20 mg, MW 6,000) in 100 μL of anhydrous
ethanol for 1 hour at 60°C under stirring. The anhydrous
ethanol was rapidly evaporated and the PTX-PEG disper-
sion was formed. These cofused samples were named as
PEG/PTX (w/w) group at 2/1, 3/1, and 4/1, respectively.
As a result, the PTX-PEG solid dispersion was formed with
PTX incorporated inside the skeleton of the hydrophilic
modifiers.
Subsequently, 45 mg of HSA was dissolved in 10 mL of
phosphate-buffered saline (PBS, 0.1 M, pH 7.4). The solution
was then mixed with the PTX-PEG solid dispersion under
magnetic stirring for 15 minutes. Then, the mixture was pre-
mixed for 10 minutes by using a high-speed disperser. Finally,
the coarse suspension was homogenized at 20,000 psi for
6 cycles. The resulting solution was filtered through a 0.22 μm
pore-sized microfiltration membrane and then lyophilized.
Cy5.5-labeled HSA and FITC-labeled PEG were used
to prepare the PTX-HSA(Cy5.5)-PEG(FITC) nanosuspen-
sion following the same method described above. To obtain
Cy5.5-labeled HSA and FITC-labeled PEG, HSA or PEG
was agitated with Cy5.5-NHS or FITC for 8 hours away from
light in the corresponding buffer solution.31–33 The mixed
solution was then dialyzed and lyophilized.
Confocal laser scanning microscopy (CLSM, LeicaTCS
SP5, Leica, Heidelberg, Germany) was used to confirm the
successful insertion of PEG into the HSA nanosuspension.
The localization of Cy5.5-labeled HSA and FITC-labeled
PEG was observed under CLSM excited at 494 and 673 nm,
and emitted at 522 and 692 nm for FITC and Cy5.5, respec-
tively. The final images were analyzed using Leica Confocal
Software.
Preparation of PTX-HSA nanosuspension
PTX-HSA nanosuspension with a size range of 100–
150 nm was prepared as reported.19 Briefly, HSA was
dissolved in the water saturated with chloroform. Mean-
while, PTX was dissolved in the consolute solutions of
chloroform and ethanol (9:1, v:v). These two solutions
were then mixed and homogenized at 20,000 psi for 9
cycles. The resulting colloid was placed under rotary
evaporation at 25°C for 15 minutes under reduced pres-
sure and lyophilized.
Characterization of PTX-PEG-HSA
nanosuspension
Determination of DL and entrapment efciency
The DL (wt%) and entrapment efficiency (EE, %) of the
PTX-PEG-HSA nanosuspension was calculated by using
the following formula:
Drug
loading (%)
Weight of PTXinnanosuspension
Weight of PTXinnan
=oosuspension
Weight of HSAfed initially
100%
+×
Entrapment
efficiency
Weight of PTXinnanosuspension
Weight o
(%) =ffPTXfedinitially ×100%
PTX concentrations were measured by HPLC (LC-2010C,
Shimadzu Corporation, Kyoto, Japan) equipped with a
Lichrospher C18 column (4.6×250 mm, 5 μm). The mobile
phase was the mixed solution of methanol and water
(75:25, v/v), while the flow rate and detection wavelength
were set at 1 mL/min and 227 nm, respectively. The sample-
injected volume was 20 μL and the column temperature was
maintained at 30°C.
Particle size and zeta potential
The lyophilized PTX-PEG-HSA and PTX-HSA powders
were redissolved by 5% (w/v) glucose solution and diluted to
5 mg/mL. The particle size and zeta potential were measured
using Malvern Zetasizer Nano-ZS90 (Malvern Instruments,
International Journal of Nanomedicine 2015:10
submit your manuscript | www.dovepress.com
Dovepress
Dovepress
7400
Yin et al
Malvern, UK). All of dynamic light scattering (DLS) mea-
surements were performed at 25°C and at a scattering angle
of 90°. The zeta potential values were calculated by the
Smoluchowski equation.
pH and osmotic pressure determination
The pH values of PTX-PEG-HSA (dissolved by 5% glucose
parenteral solution at 0.5 mg/mL of PTX) and PTX-HSA
(dissolved by 5% glucose parenteral solution at 0.5 mg/mL
of PTX) were measured using a microelectrode (Radiometer,
Copenhagen, Denmark). Determination of osmotic pressure
was conducted with an advanced freezing point osmometer
obtained from Advanced Instruments Inc (Norwood,
MA, USA).
Morphology observation
Transmission electron microscopy (TEM, H-7650, Hitachi,
Tokyo, Japan) and atomic force microscopy (AFM, Nano
Scope IIIa, Veeco, Santa Barbara, CA, USA) were employed
to observe the morphology and size distribution of PTX-PEG-
HSA nanosuspension. The samples for TEM imaging were
prepared with negative stain. Negative staining of samples
was performed as follows: a drop of sample solution was
placed onto a copper grid coated with carbon; the sample
drop was taped with a filter paper to remove surface water
and air-dried for 5 minutes, followed by the application of
0.01% phosphotungstic acid to deposit NPs on the grid. The
samples were air-dried before observation. The AFM obser-
vation was operated in tapping mode. A drop of properly
diluted micelles was placed on the surface of a clean silicon
wafer and dried under nitrogen flow at room temperature.
The AFM imaging was visualized in contact mode, using
high-resonance frequency (F0=129 kHz) pyramidal cantile-
vers with silicon probes having force constants of 20 Nm.
Scan speed was set at 2 Hz. Imaging was processed and the
widths of the particles were measured using Nanoscope
version 7.3 software.
DSC and WAXD analysis
The dispersion state of PTX in PTX-PEG and PTX-PEG-
HSA was evaluated by differential scanning calorimetry
(DSC) and wide angle X-ray diffraction (WAXD) analysis.
Analysis was conducted using an NETZSCH DSC 204
and an XD-3A powder diffraction meter (Bruker, AXS,
Karlsruhe, Germany) with CuKα-radiation, respectively.
In this study, the samples including blank HSA, PTX,
and the physical mixture of PTX and HSA were used as
controls.
Compatible stability of lyophilized PTX-
PEG-HSA
To investigate the stability of lyophilized PTX-PEG-HSA
upon resuspension, lyophilized powders of the PTX-PEG-
HSA nanosuspension were separately resuspended at
0.5 mg/mL in 0.9% sodium chloride parenteral solution and
5% glucose parenteral solution. The solutions were stored at
room temperature for 0, 8, 12, 18, or 24 hours, followed by
determination of PTX contents as described in the “Deter-
mination of DL and entrapment efficiency” section.
In vitro drug release from PTX-PEG-HSA
nanosuspension
The release profiles of PTX from the PTX-PEG-HSA nano-
suspension and the PTX-HSA nanosuspension were studied
using a simple dialysis method. The lyophilized PTX-PEG-
HSA or PTX-HSA powders containing 0.5 mg of PTX were
dispersed in 1 mL of 5% glucose solution with or without
supplement of 10% serum and placed inside a dialysis bag
(molecular weight cut off =12,000–14,000 Da). The entire
bag was immersed in a beaker containing 150 mL of PBS
containing 0.2% Tween 80 and shaken in a 37°C water bath
at 100 rpm. At predetermined time intervals, 1 mL of release
media was withdrawn and equivalent fresh release medium
was added. The amount of PTX released was determined by
HPLC analysis as described in the “Determination of DL and
entrapment efficiency” section.
In vitro cellular study
Cellular uptake
To evaluate the cellular uptake capacities of the PTX-PEG-
HSA nanosuspension, a Coumarin 6 (C6) fluorescence
probe was encapsulated into PTX-PEG-HSA and PTX-
HSA nanosuspensions accordant to our protocol for the
preparation of PTX-loaded nanosuspensions (preparation of
PTX-PEG-HSA nanosuspension). The DL of C6 in the both
suspensions was approximately 0.1%. Flow cytometry (BD
FACSCalibur, Becton Dickinson, San Jose, CA, USA) and
CLSM were used to determine the cell uptake capacities of
C6-PTX-PEG-HSA and C6-PTX-HSA. Cells were incubated
with C6-PTX-PEG-HSA or C6-PTX-HSA (C6 content:
60 μg/mL) for 1 and 4 hours at 37°C. In CLSM analysis, cells
were rinsed with PBS three times, followed by the addition
of Hoechst 33342 (10 μg/mL) to stain the cell nuclei. The
samples were then observed using an Olympus confocal
microscope (Olympus FV1000, Olympus Corporation,
Tokyo, Japan) at excitation and emission wavelengths of 488
and 530 nm, respectively. In the flow cytometry analysis, all
International Journal of Nanomedicine 2015:10 submit your manuscript | www.dovepress.com
Dovepress
Dovepress
7401
A toxic organic solvent-free technology for preparation of PTX-PEG-HSA
samples were rinsed with PBS three times to remove excess
culture media, harvested by trypsinization, and collected in
PBS to measure the fluorescence intensity.
Cytotoxicity evaluation
Approximately 100 μL of MDA-MB-231 cells at a con-
centration of 5×103 cells/well were seeded in each well
of a 96-well plate. The plate was incubated at 37°C in a
humidified atmosphere with 5% CO2. Subsequently, specific
populations of cells were treated with PTX-PEG-HSA, PTX-
HSA and Taxol® with increasing doses: 0.001–100 μg/mL
for 72 hours. After 72 hours of incubation, the MTT assay
was performed using previously reported procedures using
a microplate reader (SOFT max® PRO, Molecular Devices
Corporation, Sunnyvale, CA, USA).34 The toxicity of samples
was expressed as the inhibitory concentration at which 50%
of cell growth inhibition was obtained (IC50).
Apoptosis assay
Apoptosis of MDA-MB-231 cells induced by PTX-PEG-HSA
nanosuspensions were analyzed by flow cytometry using
AnnexinV-FITC/propidium iodide (PI) double staining
assay. MDA-MB-231 cells were cultured in 6-well plates at
a concentration of 5×104 cells/well and incubated at 37°C
in the presence of 5% CO2 for 24 h. Subsequently, the cells
were treated with 0.1 μg/mL of PTX-PEG-HSA, PTX-HSA,
or Taxol® for 24 hours, followed by three washes with cold
PBS. Cells were then harvested, washed, and resuspended
in 500 μL of PBS. Afterward, 5 μL of Annexin V-FITC and
5 μL of PI were added and incubated for 15 minutes at room
temperature in the dark. Finally, the cell suspension was
evaluated by a flow cytometry (BD FACSCalibur, Becton
Dickinson).
In vivo antitumor activity
Approximately 1×106 of MDA-MB-231 cells were inocu-
lated subcutaneously in the armpit region of athymic nude
mice. Once tumors grew to approximately 50 mm3, mice
were randomly divided into four groups and received dif-
ferent injections as follows: 1) saline (the control group,
n=5); 2) PTX-PEG-HSA (redissolved in 5% glucose, n=5);
3) PTX-HSA (redissolved in 5% glucose, n=5); 4) Taxol®
(n=5). All formulations were administered IV via the tail vein
at a PTX dose of 10 mg/kg. Treatments occurred once every
5 days for 15 days. The body weights of all mice were also
recorded, and tumor volumes were calculated as (a2×b)/2,
where a represents the smallest diameter and b the largest.
Tumors were excised from sacrificed mice after 20 days of
observation and weighed. The tumor weight inhibition rate
(IR) (%) was calculated using the following formula:
IR (%) TW TW
TW
controltest
control
=×100%
where TWcontrol and TWtest represented the mean tumor
weight of the control group and treated groups, respectively.
Thereafter, the tumor tissues were fixed in 10% of formalin
and embedded in paraffin blocks to conduct hematoxylin
and eosin (H&E) staining and terminal deoxynucleotidyl
transferase-mediated nicked labeling (TUNEL) assays.35,36
Then the stained tissue sections were observed under a light
microscope (Olympus) for histologic examination.
Statistics
Statistical evaluation was performed through a two-tailed
Student’s t-test, and one-way analysis of variance. All data
are expressed as the mean ± standard deviation (SD), unless
otherwise noted. A value of P,0.05 was considered statisti-
cally significant.
Results and discussion
Preparation and characterization of PTX-
PEG-HSA nanosuspension
Preparation of PTX-PEG-HSA solid suspension
The need to solubilize hydrophobic drugs using organic
solvents is an undesirable option from a practical standpoint
due to the potential toxicity of the solvents. In this work,
we have replaced the need for toxic organic solvents by the
solid-dispersion technology in terms of solubilizing PTX. This
method was created to further optimize the preparation of
PTX-loaded HSA nanosuspensions based on nab™. The PTX
powder was co-melted with PEG to form a solid dispersion,
which effectively improved the dissolution rate and apparent
solubility of PTX. This solid was then mixed with an HSA
solution and passed under high pressure, analogous to the
operative description used in nab™. Thus, the PTX-PEG-HSA
nanosuspension obtained from solid-dispersion technology is
free of toxic chloroform residues that limit the clinical appli-
cation of the original nab™ formulation. In other words, the
chronic toxicities of residual chloroform in Abraxane™ can be
successfully mitigated using solid-dispersion technology.
We first investigated the optimal weight ratio of PEG
to PTX for the preparation of the PTX-PEG solid suspen-
sion through co-melting. The solid dispersion technique
incorporates PTX inside the hydrophilic skeleton structure
International Journal of Nanomedicine 2015:10
submit your manuscript | www.dovepress.com
Dovepress
Dovepress
7402
Yin et al
of hydrophilic carriers, which allows detection by DSC.37,38
As shown in Figure 1A, all diagrams exhibited the endother-
mic peak of PEG around 50°C. Furthermore, only the physi-
cal mixture of PEG + PTX and the group at 2/1 (PEG/PTX,
w/w) obtained the characteristic peaks of PTX in crystalline
state around 240°C. The disappearance of characteristic
peaks of PTX in groups at 3/1 and 4/1 indicated the suc-
cessful formation of PTX-PEG solid dispersion with PTX
existing in an amorphous state. During the preparation of
the solid dispersion, the optimal pharmaceutical parameter
was defined as the minimum amount of hydrophilic carri-
ers required for the hydrophobic drugs to be sufficiently
dispersed.39 Therefore, the PTX-PEG solid dispersion
was first prepared at a weight ratio of 1:3 with respect to
PTX:PEG. This mixture was then combined with the HSA
solution and homogenized to obtain the PTX-PEG-HSA
nanosuspension.
Furthermore, a double-fluorescent label method was used
to confirm the successful attachment of PEG within the HSA-
based nanosuspension. In this method, HSA and PEG were
first tagged with Cy5.5 and FITC, respectively. Thereafter,
the PTX-HSA(Cy5.5)-PEG(FITC) nanosuspension was
prepared following the procedures of PTX-PEG-HSA manu-
facture. The formation of PTX-HSA(Cy5.5)-PEG(FITC)
nanosuspension was evaluated visually under a CLSM. As
shown in Figure 1B, the merged green/red fluorescence
(indicated by white arrows) of PTX-HSA(Cy5.5)-PEG(FITC)
nanosuspensions was obtained via confocal microscopy
imaging. This result directly demonstrated that PTX-
PEG-HSA nanosuspensions could be formed through our
toxic organic solvent-free technology (with successful PEG
insertion). H-bonding network between PEG and vicinal
amino acids of HSA surrounding may play a major role in
the PEG/HSA complexation.40
2QVHW&
2QVHW&
P P P
),7&
$DF
EG
%&\ 0HUJHG
2QVHW&
2QVHW&
2QVHW& 2QVHW& 2QVHW&
2QVHW&
Figure 1 (A) DSC thermograms of (a) physical mixture of PEG + PTX; (b) the PTX-PEG solid dispersion prepared at weight ratio 2/1 (PEG/PTX); (c) the PTX-PEG solid
dispersion prepared at weight ratio 3/1 (PEG/PTX); (d) the PTX-PEG solid dispersion prepared at weight ratio 4/1 (PEG/PTX). (B) Confocal microscopy of PTX-HSA(Cy5.5)-
PEG(FITC).
Notes: For each panel, images from left to right show the uorescence of PEG-FITC (green), HSA-Cy5.5 (red), and the overlays of PTX-HSA(Cy5.5)-PEG(FITC) with
uorescence of PEG-FITC and HSA-Cy5.5 indicated by white arrows.
Abbreviations: DSC, differential scanning calorimetry; PEG, polyethylene glycol; PTX, paclitaxel; FITC, uorescein isothiocyanate; HSA, human serum albumin.
International Journal of Nanomedicine 2015:10 submit your manuscript | www.dovepress.com
Dovepress
Dovepress
7403
A toxic organic solvent-free technology for preparation of PTX-PEG-HSA
Determination of DL and EE
All of the physicochemical characteristics of PTX-loaded
nanosuspensions are listed in Table 1. First, DL capability
was determined using HPLC. The DL (wt%) and EE (%)
of PTX-PEG-HSA was 9.1±0.2 wt% and 90.5%±0.6%,
respectively. PTX-HSA obtained a DL and an EE of
9.2±0.1 wt% and 91.4%±0.5%, respectively. The similar
DL and EE observed in these two HSA-based nanosuspen-
sions indicated that the unique drug dispersion by PEG did
not affect the specific binding interactions between PTX
and HSA.
Particle size and zeta potential evaluation
The particle size and zeta potential of PTX-PEG-HSA and
PTX-HSA were measured by DLS. As listed in Table 1,
the effective diameters of PTX-PEG-HSA and PTX-HSA
were 123.0 and 137.2 nm with polydispersity indices (PDI)
of 0.11 and 0.14, respectively, implying a narrow size
distribution. In addition, the particle surface of PTX nano-
suspensions were negatively charged with zeta potential of
approximately -43.6 mV for PTX-PEG-HSA and -39.9 mV
for PTX-HSA. The mean particle size distribution usually
affects the formulation stability, pharmacokinetics, and in
vivo tissue distribution of nanosuspension systems.41 As
compared with PTX-HSA, PTX-PEG-HSA nanosuspen-
sions shared a similar zeta potential but had a smaller mean
diameter and are less likely to be nonselectively recognized
by the RES. This decrease in RES uptake, therefore, allows
the particles to achieve long-circulation times in blood by
avoiding RES clearance.42 Additionally, the EPR effect was
reported to be a significant factor when the size of NPs was
approximately 100–200 nm.43 Furthermore, the relatively
high zeta potential provides a repelling force between the
particles. This electrostatic repulsion acts to the formulation’s
advantage to prevent undesired aggregation or precipitation
and thus, improves the formulation’s stability.44 Therefore, it
is reasonable to conclude that PTX-loaded nanosuspensions
with small diameters (123.0 nm) and negatively charged
zeta potentials (-43.6 mV) are suitable for targeted tumor
therapy through IV injection due to the reasons that have
just been listed.
pH and osmotic pressure determination
Both the PTX-PEG-HSA and PTX-HSA nanosuspensions
exhibited pH values around 7.4 and osmotic pressures in the
range of 300–340 mOsm/kg, which agrees with the optimal
measurements for IV injections.
Morphology observation
TEM and AFM were used to visualize the size and morphology
of PTX-PEG-HSA particles. As shown in Figure 2, the mor-
phology of both the nanosuspensions was spherical, and the
particles were highly dispersed with diameters approximately
100 nm. It was also noted that the particle size observed by
TEM and AFM is smaller than that determined by DLS.
In the DLS measurement, the hydrated dynamic diameter
of NPs was measured, ie, the colloidal hydrate layer is
calculated as one part of “diameter”. In the microscopy
determination, the colloidal hydrate layer is dried and the
observed true diameter decreased. Therefore, it makes sense
that the measurements observed with dehydrated samples in
TEM and AFM would be smaller than those determined with
hydrated samples during DLS.
DSC and WAXD analysis
To investigate the physical state of PTX in the nanosus-
pensions, DSC and WAXD analysis were conducted for
PTX-PEG-HSA with PEG, HSA, PTX, a physical mixture
of HSA + PTX, and a physical mixture of HSA + PTX-PEG
as controls. DSC diagrams of the six samples are shown in
Figure 3A. As illustrated in Figure 3A(a), the endothermic
peak at 217.3°C and exothermic peak at 244.5°C were
characteristic peaks of PTX in crystalline state, while the
exothermic peak at 208.9°C in Figure 3A(b) represented
the peak of decomposed HSA. Figure 3A(c) exhibited the
endothermic peak of PEG at around 50°C. Additionally, an
exothermic peak at 245.8°C was seen for the physical mix-
ture of HSA + PTX (Figure 3A(d)), but was nearly absent
for HSA + PTX-PEG (Figure 3A(e)) and PTX-PEG-HSA
nanosuspensions (Figure 3A(f)). All results conclude that
the PTX were physically encapsulated in these nanosus-
pensions with reduced crystallinity. The results of WAXD
analysis in Figure 3B further confirmed the existing state
Table 1 Physicochemical characteristics of PTX-PEG-HSA and PTX-HSA nanosuspension
Groups DL (wt%)aEE (%)aSize (nm) PDIbZeta potential (mV) pH OP (mOsm/kg)c
PTX-PEG-HSA 9.1±0.2 90.5±0.6 123.0±2.1 0.11 -43.6±1.8 7.3±0.1 322±4.2
PTX-HSA 9.2±0.1 91.4±0.5 137.2±1.9 0.14 -39.9±3.2 7.3±0.2 325±6.2
Notes: aMean diameters of the nanosuspensions, detected by DLS. bPolydispersity index (PDI) of the nanosuspensions, detected by DLS. cOsmotic pressure (OP) of the
nanosuspensions.
Abbreviations: PTX, paclitaxel; PEG, polyethylene glycol; HSA, human serum albumin; DLS, dynamic light scattering; DL, drug loading; EE, encapsulation efciency.
International Journal of Nanomedicine 2015:10
submit your manuscript | www.dovepress.com
Dovepress
Dovepress
7404
Yin et al
D
QP QP
QP
 P
+HLJKW
QP
$
%
EF
Figure 2 (A) TEM images of free PTX (a), PTX-PEG-HSA (b), and PTX-HSA (c), and (B) the AFM images of PTX-PEG-HSA.
Abbreviations: TEM, transmission electron microscopy; PTX, paclitaxel; PEG, polyethylene glycol; HSA, human serum albumin; AFM, atomic force microscopy.
2QVHW&
2QVHW&
2QVHW&
2QVHW&
2QVHW&
2QVHW&
2QVHW&
2QVHW&
2QVHW&
7HPSHUDWXUH& θ 
$
D
E
F
G
H
I
%
D
E
F
G
H
I
           
2QVHW&
2QVHW&
2QVHW&
2QVHW&
Figure 3 (A) DSC thermograms of (a) PTX; (b) lyophilized HSA; (c) PEG; (d) a physical mixture of HSA + PTX; (e) a physical mixture of HSA + PTX-PEG; (f) PTX-PEG-HSA.
(B) Powder X-ray diffraction patterns for (a) PTX; (b) lyophilized HSA; (c) PEG; (d) a physical mixture of HSA + PTX; (e) a physical mixture of HSA + PTX-PEG;
(f) PTX-PEG-HSA.
Abbreviations: DSC, differential scanning calorimetry; PTX, paclitaxel; PEG, polyethylene glycol; HSA, human serum albumin.
International Journal of Nanomedicine 2015:10 submit your manuscript | www.dovepress.com
Dovepress
Dovepress
7405
A toxic organic solvent-free technology for preparation of PTX-PEG-HSA
of PTX loaded in PTX-PEG-HSA nanosuspensions. As
shown in Figure 3B(a), PTX had three intense peaks at 5°,
9°, and 12° and small peaks at 15°–30°. The HSA gave a
broad and weak peak at 15°–30°. The typical crystal peaks
of PTX and HSA were all present in the WAXD diagram of
a physical mixture of PTX + HSA. However, the PTX-PEG-
HSA nanosuspensions possessed negligible characteristic
peaks of PTX and exhibited one obvious peak, similar to
that of free HSA, which also demonstrated that the PTX
were encapsulated in nanosuspensions with a significantly
reduced crystalline state.45
Compatible stability for clinical
application
The lyophilized powders of this formulation are usually
redissolved by 0.9% sodium chloride or 5% glucose injection
for clinical application. Therefore, the stability and compat-
ibility of resuspensions are a crucial criterion for formulation
design. In this study, PTX-PEG-HSA redissolved in 0.9%
sodium chloride or 5% glucose injection was analyzed for the
amount of unincorporated PTX and particle size. As listed in
Table 2, there was no significant change of the two indexes
within 24 hours, suggesting that both the 0.9% sodium chlo-
ride and 5% glucose injections can be used for resuspending
the PTX-PEG-HSA from lyophilized powders.
In vitro drug release
Drug release rate was determined to examine whether a
burst release pattern was present, or whether the drug could
be slowly dissipated from the polymer matrix for exerting
potency. Therefore, PTX-PEG-HSA and PTX-HSA nano-
suspensions were incubated in PBS at pH 7.4, and the PTX
release profiles were determined. As shown in Figure 4A,
both PTX-PEG-HSA and PTX-HSA nanosuspensions
exhibited steady and continual release profiles with no initial
burst release. The results suggested that PTX molecules were
indeed encapsulated into the hydrophobic micropockets of
HSA and not lodged on the particle surface. This finding
agreed with the results of DSC and WAXD analysis. To
simulate the in vivo drug release, the release evaluation of
PTX-PEG-HSA and PTX-HSA in 10% serum supplemented
medium was also conducted. The results shown in Figure 4B
exhibited a similar slow drug release behavior of PTX-PEG-
HSA within 24 hours to that obtained in PBS solution and
an increased drug release from PTX-HSA as compared with
that in PBS solution. These results indicated that PEG in
PTX-PEG-HSA could protect NPs from protein interaction
and the PTX-PEG-HSA could achieve a less drug preleakage
during blood circulation, therefore improving the circulative
stability and also mitigating side effects. Another point worth
noting is that the amount of PTX released from PTX-HSA
was lower than that from PTX-PEG-HSA over the same time
intervals in PBS without serum. Within the first 4 days, only
approximately 35% of PTX was released from PTX-HSA,
while the release amount in PTX-PEG-HSA group reached
up to 53%. This result may be attributed to the insertion of
PEG within PTX-PEG-HSA nanosuspensions that furthers
drug diffusion. Finally, the cumulative PTX release per-
centage of both preparations reached 80% after 10 days in
serum-free media, indicating successful PTX release from
designed nanosuspensions.
In vitro cellular studies
Cellular uptake and intracellular distribution
With confirmed physicochemical properties of PTX-PEG-
HSA nanosuspension, investigation into the cellular uptake
behavior of the nanosuspension using a C6 as fluorescent
marker was conducted. The results from flow cytometry
analysis are shown in Figure 5A. MDA-MB-231 cells incu-
bated with the C6-PTX-PEG-HSA and C6-PTX-HSA both
exhibited fluorescence of C6 at 1 and 4 hours, indicating
a time-dependent cellular internalization mechanism.
Meanwhile, the fluorescence intensity in the PTX-PEG-
HSA group was approximately equal to that in PTX-HSA
group at 1 and 4 h, respectively. This result demonstrates
that the PEGylation did not adversely affect the cell uptake
of PTX-PEG-HSA, which could be mediated through the
60 KD glycoprotein (gp60) or SPARC receptor on the cell
membrane.46 The highly effective cellular internalization of
PTX-loaded nanosuspensions was also confirmed via CLSM
analysis. The time dependence of endocytosis and strength
of intracellular fluorescence is shown in Figure 5B and is in
agreement with the results from flow cytometry analysis. In
addition, the internalized fluorescence of C6 was distributed
Table 2 The compatible stability of PTX-PEG-HSA with 5%
glucose and 0.9% sodium chloride injections (n=3)
Time (h) 5% glucose 0.9% sodium chloride
Size (nm) PTX
content (%)
Size (nm) PTX
content (%)
0122.5±2.9 100.0±3.2 124.5±2.2 100.0±2.1
8124.7±7.9 100.1±5.1 125.2±4.6 100.8±3.0
12 125.8±6.1 101.1±1.2 128.4±5.9 99.7±7.7
18 128.4±9.2 100.8±2.5 126.7±8.3 101.3±6.1
24 126.9±3.2 99.1±1.9 127.4±6.5 101.2±3.8
Abbreviations: PTX, paclitaxel; PEG, polyethylene glycol; HSA, human serum
albumin.
International Journal of Nanomedicine 2015:10
submit your manuscript | www.dovepress.com
Dovepress
Dovepress
7406
Yin et al
Figure 4 (A) In vitro release of PTX from PTX-PEG-HSA and PTX-HSA in media of PBS at pH 7.4; (B) PTX release proles of PTX-HSA and PTX-PEG-HSA in 10% serum
supplemented media at pH 7.4.
Note: Data are presented as the mean ± SD (n=3).
Abbreviations: PTX, paclitaxel; PEG, polyethylene glycol; HSA, human serum albumin; PBS, phosphate-buffered saline; SD, standard deviation.
throughout the cytoplasm, the optimal location for PTX to
induce cytotoxicity. Therefore, all the results here indicated
that our designed nanosuspension could be quickly taken into
tumor cells and reside in the preferred intracellular location
for inducing optimal cytotoxicity.
Cytotoxicity and apoptosis assay
To further investigate whether the highly efficient intra-
cellular PTX delivery platform (PTX-PEG-HSA) could
achieve strong cytotoxicity against tumor cells, an MTT
assay of PTX-PEG-HSA against MDA-MB-231 cells was
performed using PEG-HSA and Taxol® as controls. After
24 hours of incubation, Taxol® had significantly higher IC50
values against MDA-MB-231 cells than PTX-HSA and PTX-
PEG-HSA (P,0.001, Figure 6A). This phenomenon could
be attributed to a number of reasons. First, Taxol® primarily
utilizes passive diffusion to enter the cell, and this is solely
dependent on the concentration gradient. On the other hand,
the two HSA-based nanosuspensions could be transported
into tumor cells through active binding of HSA to the gp60
or SPARC receptor on cell membrane. Furthermore, PTX in
the HSA-based nanosuspensions accumulated in the tumor
cells, while free PTX was rapidly secreted back outside the
cells.47 In addition, the IC50 of the PTX-PEG-HSA group was
1.76-fold lower than that of PTX-HSA group. As stated, the
higher cytotoxicity of PTX-PEG-HSA on tumor cells may be
attributed to its faster drug release behavior, which has been
confirmed by the in vitro drug release experiment.
The significant inhibitory effect of PTX-PEG-HSA
on MDA-MB-231 cells was also demonstrated by an
AnnexinV-FITC/PI double staining assay, and cell apoptosis
was measured via flow cytometry. The representative images
obtained from flow cytometry shown in Figure 6C mirrored
results from the MTT assay. In detail (Figure 6B), the early
apoptosis (Annexin V+/PI-) value of MDA-MB-231 cells
treated with PTX-PEG-HSA, PTX-HSA, and Taxol® was
12.9%, 10.6%, and 10.6%, respectively. The percentage of
late apoptosis/necrosis cells (AnnexinV+/PI+) was 10.3%,
6.69%, and 3.45%, respectively. These results indicated
that PTX-PEG-HSA could induce elevated apoptosis over
PTX-HSA and Taxol®.
In vivo antitumor activity
To investigate the in vivo antitumor potential of PTX-PEG-
HSA nanosuspensions, the antitumor efficacy was further
evaluated on BALB/c nude mice bearing MDA-MB-231
xenografts with PTX-HSA and Taxol® as controls. The
changes of tumor volume over 20 days of treatment and
the IR (%) values of these PTX formulations calculated on
the basis of tumor weight are depicted in Figure 7A and B,
respectively. As indicated, all PTX formulations exhibited
effective inhibition on tumor growth when compared with
saline group. However, the PTX-PEG-HSA nanosuspension
achieved the strongest tumor inhibition of all four groups,
with an IR value of 89.24%, followed by Taxol® (79.96%)
and PTX-HSA (75.08%).
Furthermore, the remarkable antitumor efficacy of the
PTX-PEG-HSA nanosuspensions is also well supported by
H&E histological evaluation and TUNEL assay. Unlike the
tumors in saline control group with histologic characteristics
International Journal of Nanomedicine 2015:10 submit your manuscript | www.dovepress.com
Dovepress
Dovepress
7407
A toxic organic solvent-free technology for preparation of PTX-PEG-HSA
&37;+6$IRUKRXU
P P
P P
&37;+6$IRUKRXUV
0HUJHGIOXRUHVFHQFHRI&DQG+RHFKVW
&37;3(*+6$IRUKRXU &37;3(*+6$IRUKRXUV
$
%


0HDQIOXRUHVFHQFHLQWHQVLW\DX

7LPHKRXU
%ODQNFRQWURO
&37;+6$
&37;3(*+6$
Figure 5 Flow cytometry (A) and CLSM (B) measurements of the intracellular uptake of C6-PTX-PEG-HSA and C6-PTX-HSA at 1 and 4 hours in MDA-MB-231 cells.
Note: Results were expressed as the mean ± SD (n=5).
Abbreviations: CLSM, confocal laser scanning microscopy; PTX, paclitaxel; PEG, polyethylene glycol; HSA, human serum albumin; SD, standard deviation; C6, Coumarin 6.
including hyper chromatic nuclei, scant cytoplasm, and
closely arranged tumor cells, the tumors of mice treated
with PTX-PEG-HSA, PTX-HSA or Taxol® all exhibited
cell shrinkage and intercellular blank (Figure 8). Yet,
tumors treated with PTX-PEG-HSA exhibited the great-
est degree of apoptosis observed through TUNEL assay
and the lowest tumor cellularity. The enhanced efficacy of
these two HSA-based nanosuspensions over Taxol® might
International Journal of Nanomedicine 2015:10
submit your manuscript | www.dovepress.com
Dovepress
Dovepress
7408
Yin et al




&HOOYLDELOLW\
$SRSWRVLVFHOOFRXQWV
&RQFHQWUDWLRQJP/


3,
$QQH[LQ),7&

4

4

4

4










3,
$QQH[LQ),7&

4

4

4

4










3,
$QQH[LQ),7&

4

4

4

4










3,
$QQH[LQ),7&

4

4

4

4














     
37;3(*+6$,& QJP/
37;+6$,& QJP/
7D[RO,& QJP/
6DOLQH
37;+6$ 37;3(*
+6$
6DOLQH
37;+6$
37;3(*+6$
7D[RO
7D[RO
$
&DE
FG
%
Figure 6 Cytotoxicity and apoptosis assay of MDA-MB-231 cells incubation with different formulations.
Notes: (A) Viability of MDA-MB-231 cells after incubation with different formulations at different concentrations after 24 hours. The values are represented as mean ± SD
(n=5). *P,0.01 versus Taxol®, **P,0.05 versus PTX-HSA. (B) Different counts of apoptosis and necrotic cells after treatment with different formulations. *P,0.01.
(C) Representative results of Annexin V-FITC/PI double labeling assay. Cells were treated with saline (a), PTX-PEG-HSA (b), PTX-HSA (c), or Taxol® (d) for 24 hours.
Early apoptosis (AnnexinV+/PI-), late apoptosis and necrotic cells (AnnexinV+/PI+) are shown.
Abbreviations: PTX, paclitaxel; PEG, polyethylene glycol; HSA, human serum albumin; SD, standard deviation; PI, propidium iodide; FITC, uorescein isothiocyanate.
International Journal of Nanomedicine 2015:10 submit your manuscript | www.dovepress.com
Dovepress
Dovepress
7409
A toxic organic solvent-free technology for preparation of PTX-PEG-HSA





'D\V
$%
7XPRUYROXPHPP
7XPRUZHLJKWJ
  







6DOLQH
37;+6$
37;3(*+6$
7D[RO
6DOLQH
37;+6$,5 
37;3(*+6$,5 
7D[RO,5 
6DOLQH 37;+6$ 37;3(*
+6$
7D[RO
Figure 7 Tumor growth inhibition (A), tumor weight (B), and the percentage of tumor weight IR observed in MDA-MB-231 tumor-bearing nude mice treated with saline,
Taxol®, PTX-HSA, and PTX-PEG-HSA (n=5). *P,0.01, **P,0.05.
Abbreviations: IR, inhibition rate; PTX, paclitaxel; HSA, human serum albumin; PEG, polyethylene glycol.
6DOLQH
+(781(/
37;+6$ 37;3(*+6$7D[RO
Figure 8 H&E staining and TUNEL analysis of tissue sections of tumor excised from the mice 20 days after treatment.
Abbreviations: PTX, paclitaxel; HSA, human serum albumin; PEG, polyethylene glycol; H&E, hematoxylin and eosin; TUNEL, terminal deoxynucleotidyl transferase-
mediated nicked labeling.
be explained by the combination of EPR effect and HSA
binding receptor-mediated cellular internalization, which
significantly improved PTX accumulation in tumors.48,49
Moreover, the additional PEGylation of PTX-PEG-HSA
prolonged the in vivo half-life of particles in circulation
through its unique “stealth function” to prevent RES rec-
ognition, thereby leading to a further increased target dis-
tribution of PTX into tumors. Finally, the accelerated drug
release behavior of PTX-PEG-HSA over PTX-HSA further
enhanced the induction of apoptosis. Consequently, a more
effective anticancer activity was achieved by PTX-PEG-
HSA compared with PTX-HSA.
It is known that in vivo toxicity evaluation is essential for
systemic drug delivery. Therefore, we investigated animal
behavior and body weight during formulation treatments.
First, all mice in PTX-PEG-HSA and PTX-HSA groups
showed normal behavior during the entire treatment, while
the mice treated with Taxol® frequently demonstrated
decreased activity and appetite. Additionally, mice in the
Taxol® group exhibited significant weight loss (Figure 9). In
contrast, mice receiving HSA formulation-based treatments
revealed negligible changes in body weight. The obvious side
effects of Taxol® treatment have already been well explained
by the dose-limiting toxicity of the nonionic surfactant
International Journal of Nanomedicine 2015:10
submit your manuscript | www.dovepress.com
Dovepress
Dovepress
7410
Yin et al
Cremophor® EL alongside the nonspecific distribution of
PTX.50 The marked reduction of systemic toxicity achieved
by HSA nanosuspensions was mainly due to the effective
encapsulation of PTX into the HSA-based particles, which
avoided Cremophor EL-related side effects. Furthermore, the
well-defined selective accumulation of HSA nanosuspensions
in tumor sites through both the EPR and protein-receptor-
mediated endocytosis enhanced the tumor specificity and
reduced systemic cytotoxicity.51 Additionally, the HSA-based
particles demonstrated decreased leakage of PTX into the
bloodstream during circulation as compared with Taxol®,
which further mitigated side effects.52
Conclusion
The solid-dispersion technology has allowed the formulation
of PTX-PEG-HSA, a powerful nanosuspension-based deliv-
ery system for PTX that circumvents the need for organic
solvents and Cremophor EL. PTX molecules were success-
fully dispersed by PEG through co-melting, followed by
combination, and homogenized with HSA solution. Particles
were confirmed to be of a small size (123.0 nm) with smooth
spherical morphology. Furthermore, the PTX molecules in
PTX-PEG-HSA were dispersed in a molecular or amorphous
state. The stability of this formulation was evident through
the steady, continued drug release behavior without an initial
burst release effect, which suggested the promising potential
of PTX-PEG-HSA as a safe systemic PTX delivery platform.
The high cellular internalization of PTX-PEG-HSA as
demonstrated by flow cytometry and CLSM analysis shows
that stable NPs could still be internalized into the cell. In
addition, the strong cytotoxicity of PTX-PEG-HSA against
MDA-MB-231 cells indicates that encapsulated PTX can
be released in the intracellular environment for significant
antitumor efficacy. More importantly, in vivo antitumor
investigation on nude mice bearing MDA-MB-231 cancer
xenografts further confirmed that PTX-PEG-HSA achieved
improved anticancer activity and reduced systemic toxic-
ity over PTX-HSA and Taxol®. The use of PEG and HSA
receptor-mediated targeting has allowed for prolonged blood
circulation and significant increases in tumor uptake, all of
which have translated to enhanced antitumor efficacy with
decreased collateral damage to healthy tissues. Thus, PTX-
PEG-HSA without dependence on toxic organic solvents
proves a greater alternative to commonly accepted PTX
delivery systems. Future work in this direction should focus
on in vivo pharmacokinetics and biodistribution of PTX-
PEG-HSA on tumor-bearing mice, as well as evaluation
of the safety and activity of the delivery system in a more
clinically accurate model.
Acknowledgments
This work was supported by the project of the National
Natural Science Foundation of China (number 81102397),
the Natural Science Foundation of Jiangsu Province (number
BK2012761), Qing Lan Project of Jiangsu Province (number
02432009), Innovative Project for Graduate Cultivation
of Jiangsu Province (number CXZZ11-0807), the Project
Program of State Key Laboratory of Natural Medicines,
China Pharmaceutical University (numbers JKGQ201107,
JKPZ2013004), Key New Drug Innovation Project from
the Ministry of Science and Technology of China (number
2009ZX09310004), Fostering Plan of University Scientific
and Technological Innovation Team of Jiangsu Qing Lan
Project (2014), and National Basic Research Program of
China (973 Program, number 2009CB903300).
Disclosure
The authors report no conflicts of interest in this work.
References
1. Jose Merlin JP, Venkadesh B, Hussain R, et al. Paclitaxel loaded poly-d,
l-lactide-co-glycolide nanoparticles: enhanced anticancer effect in non-small
cell lung carcinoma cell line. Biomed Prevent Nutr. 2013;3(1):1–9.
2. Tao Y, Han J, Wang X, Dou H. Nano-formulation of paclitaxel by
vitamin E succinate functionalized pluronic micelles for enhanced encap-
sulation, stability and cytotoxicity. Colloid Surf B Biointerfaces. 2013;
102:604–610.








'D\V
%RG\ZHLJKWJ
 
6DOLQH
37;+6$
7D[RO
37;3(*+6$
Figure 9 Body weight change of nude mice bearing MDA-MB-231 cells after IV
injection of different PTX formulations.
Note: *P,0.01 versus the saline group.
Abbreviations: PTX, paclitaxel; HSA, human serum albumin; PEG, polyethylene
glycol; IV, intravenous.
International Journal of Nanomedicine 2015:10 submit your manuscript | www.dovepress.com
Dovepress
Dovepress
7411
A toxic organic solvent-free technology for preparation of PTX-PEG-HSA
3. Xin H, Chen L, Gu J, et al. Enhanced anti-glioblastoma efficacy by
PTX-loaded PEGylated poly(ε-caprolactone) nanoparticles: in vitro
and in vivo evaluation. Int J Pharm. 2010;402(1–2):238–247.
4. Sun Y, Yu B, Wang G, et al. Enhanced antitumor efficacy of vitamin E
TPGS-emulsified PLGA nanoparticles for delivery of paclitaxel. Colloid
Surf B Biointerfaces. 2014;123:716–723.
5. Sharma S, Verma A, Teja BV, Shukla P, Mishra PR. Development of
stabilized Paclitaxel nanocrystals: In-vitro and in-vivo efficacy studies.
Eur J Pharm Sci. 2015;69:51–60.
6. Pawar AP, Vinugala D, Bothiraja C. Nanocochleates derived from
nanoliposomes for paclitaxel oral use: preparation, characterization,
in vitro anticancer testing, bioavailability and biodistribution study in
rats. Biomed Pharmacother. 2014;3502:1–9.
7. Yan F, Li L, Deng Z, et al. Paclitaxel-liposome-microbubble complexes
as ultrasound-triggered therapeutic drug delivery carriers. J Control
Release. 2013;166(3):246–255.
8. Lundberg BB, Risovic V, Ramaswamy M, Wasan KM. A lipophilic
paclitaxel derivative incorporated in a lipid emulsion for parenteral
administration. J Controll Release. 2003;86(1):93–100.
9. Rapoport N, Gupta R, Kim YS, O’Neill BE. Polymeric micelles and
nanoemulsions as tumor-targeted drug carriers: insight through intravital
imaging. J Control Release. 2015;206:153–160.
10. Bouquet W, Ceelen W, Fritzinger B, et al. Paclitaxel/beta-cyclodextrin
complexes for hyperthermic peritoneal perfusion formulation and
stability. Eur J Pharm Biopharm. 2007;66(3):391–397.
11. Chun C, Lee SM, Kim SY, Yang HK, Song SC. Thermosensitive
poly(organophosphazene)-paclitaxel conjugate gels for antitumor
applications. Biomaterials. 2009;30(12):2349–2360.
12. Gao L, Liu G, Kang J, et al. Paclitaxel nanosuspensions coated with
P-gp inhibitory surfactants: I. Acute toxicity and pharmacokinetics
studies. Colloids Surf B Biointerfaces. 2013;111:277–281.
13. Schleich N, Sibret P, Danhier P, et al. Dual anticancer drug/
superparamagnetic iron oxide-loaded PLGA-based nanoparticles for
cancer therapy and magnetic resonance imaging. Int J Pharm. 2013;
447(1–2):94–101.
14. Chin WW, Parmentier J, Widzinski M, Tan EH, Gokhale R. A brief
literature and patent review of nanosuspensions to a final drug product.
J Pharm Sci. 2014;103(10):2980–2999.
15. Langer K, Anhorn MG, Steinhauser I, et al. Human serum albumin
(HSA) nanoparticles: reproducibility of preparation process and kinetics
of enzymatic degradation. Int J Pharm. 2008;347(1–2):109–117.
16. Ranjbar S, Shokoohinia Y, Ghobadi S, et al. Studies of the interaction
between isoimperatorin and human serum albumin by multispectro-
scopic method: identification of possible binding site of the compound
using esterase activity of the protein. Scientific World J. 2013;2013:
305081.
17. Chatpun S, Nacharaju P, Cabrales P. Improving cardiac function with
new-generation plasma volume expanders. Am J Emerg Med. 2013;
31(1):54–63.
18. Son S, Song S, Lee SJ, et al. Self-crosslinked human serum albumin
nanocarriers for systemic delivery of polymerized siRNA to tumors.
Biomaterials. 2013;34(37):9475–9485.
19. Li C, Li Y, Gao Y, et al. Direct comparison of two albumin-based
paclitaxel-loaded nanoparticle formulations: is the crosslinked version
more advantageous? Int J Pharm. 2014;468(1–2):15–25.
20. Ibrahim N, Ibrahim H, Dormoi J, et al. Albumin-bound nanoparticles
of practically water-insoluble antimalarial lead greatly enhance its
efficacy. Int J Pharm. 2014;464(1–2):214–224.
21. Crisante F, Francolini I, Bellusci M, Martinelli A, D’Ilario L, Piozzi A.
Antibiotic delivery polyurethanes containing albumin and polyallylam-
ine nanoparticles. Eur J Pharm Sci. 2009;36(4–5):555–564.
22. Yu S, Yao P, Jiang M, Zhang G. Nanogels prepared by self-assembly
of oppositely charged globular proteins. Biopolymers. 2006;83(2):
148–158.
23. Langer K, Balthasar S, Vogel V, Dinauer N, von Briesen H, Schubert D.
Optimization of the preparation process for human serum albumin
(HSA) nanoparticles. Int J Pharm. 2003;257(1–2):169–180.
24. Elzoghby AO, Samy WM, Elgindy NA. Albumin-based nanoparticles
as potential controlled release drug delivery systems. J Control Release.
2012;157(2):168–182.
25. Lluch A, Alvarez I, Munoz M, Segui MA, Tusquets I, Garcia-Estevez L.
Treatment innovations for metastatic breast cancer: nanoparticle
albumin-bound (NAB) technology targeted to tumors. Crit Rev Oncol
Hematol. 2014;89(1):62–72.
26. Gradishar WJ. Albumin-bound paclitaxel: a next-generation taxane.
Expert Opin Pharmacother. 2006;7(8):1041–1053.
27. Catauro M, Bollino F, Papale F, Gallicchio M, Pacifico S. Influence of
the polymer amount on bioactivity and biocompatibility of SiO2/PEG
hybrid materials synthesized by sol-gel technique. Mat Sci Eng C Mater
Biol Appl. 2015;48:548–555.
28. Qiu L, Zheng C, Jin Y, Zhu K. Polymeric micelles as nanocarriers for
drug delivery. Expert Opin Ther Patents. 2007;17(7):819–830.
29. Danhier F, Lecouturier N, Vroman B, et al. Paclitaxel-loaded PEGylated
PLGA-based nanoparticles: in vitro and in vivo evaluation. J Control
Release. 2009;133(1):11–17.
30. Cui F, Li Y, Zhou S, et al. A comparative in vitro evaluation of self-
assembled PTX-PLA and PTX-MPEG-PLA nanoparticles. Nanoscale
Res Lett. 2013;8(1):301–308.
31. Hahnenkamp A, Alsibai W, Bremer C, Holtke C. Optimizing the bio-
availability of small molecular optical imaging probes by conjugation
to an albumin affinity tag. J Control Release. 2014;186:32–40.
32. Zhong Y, Yang W, Sun H, et al. Ligand-directed reduction-sensitive
shell-sheddable biodegradable micelles actively deliver doxorubicin
into the nuclei of target cancer cells. Biomacromolecules. 2013;14(10):
3723–3730.
33. Xie J, Chen K, Huang J, et al. PET/NIRF/MRI triple functional iron
oxide nanoparticles. Biomaterials. 2010;31(11):3016–3022.
34. Wang H, Sun X, Gao F, Zhong B, Zhang YH, Sun Z. Effect of ulinastatin
on growth inhibition, apoptosis of breast carcinoma cells is related to
a decrease in signal conduction of JNk-2 and NF-kappaB. J Exp Clin
Cancer Res. 2012;31:1–13.
35. Pelzer ES, Harris JE, Allan JA, et al. TUNEL analysis of DNA frag-
mentation in mouse unfertilized oocytes: the effect of microorganisms
within human follicular fluid collected during IVF cycles. J Reprod
Immunol. 2013;99(1–2):69–79.
36. Wu D, Zheng Y, Hu X, Fan Z, Jing X. Anti-tumor activity of folate
targeted biodegradable polymer-paclitaxel conjugate micelles on
EMT-6 breast cancer model. Mater Sci Eng C Mater Biol Appl. 2015;53:
68–75.
37. Dang Y, Zhu C. Genomic study of the absorption mechanism of can-
tharidin and its solid dispersion. Colloid Surf A Physicochem Eng Asp.
2012;415:295–301.
38. Liu X, Lu M, Guo Z, Huang L, Feng X, Wu C. Improving the chemical
stability of amorphous solid dispersion with cocrystal technique by hot
melt extrusion. Pharm Res. 2012;29(3):806–817.
39. Lima AA, Soares-Sobrinho JL, Silva JL, et al. The use of solid disper-
sion systems in hydrophilic carriers to increase benzonidazole solubility.
J Pharm Sci. 2011;100(6):2443–2451.
40. Bekale L, Agudelo D, Tajmir-Riahi HA. The role of polymer size and
hydrophobic end-group in PEG-protein interaction. Colloid Surf B
Biointerfaces. 2015;130:141–148.
41. Yadav SK, Mishra S, Mishra B. Eudragit-based nanosuspension of
poorly water-soluble drug: formulation and in vitro-in vivo evaluation.
AAPS Pharm Sci Tech. 2012;13(4):1031–1044.
42. Guo S, Huang L. Nanoparticles escaping RES and endosome: challenges
for siRNA delivery for cancer therapy. J Nanomater. 2011;2011:11.
43. Maeda H, Bharate GY, Daruwalla J. Polymeric drugs for efficient tumor-
targeted drug delivery based on EPR-effect. Eur J Pharm Biopharm.
2009;71(3):409–419.
44. Wu L, Zhang J, Watanabe W. Physical and chemical stability of drug
nanoparticles. Adv Drug Deliv Rev. 2011;63(6):456–469.
45. Huo M, Zhang Y, Zhou J, et al. Synthesis and characterization of low-
toxic amphiphilic chitosan derivatives and their application as micelle
carrier for antitumor drug. Int J Pharm. 2010;394(1–2):162–173.
International Journal of Nanomedicine
Publish your work in this journal
Submit your manuscript here: http://www.dovepress.com/international-journal-of-nanomedicine-journal
The International Journal of Nanomedicine is an international, peer-
reviewed journal focusing on the application of nanotechnology
in diagnostics, therapeutics, and drug delivery systems throughout
the biomedical field. This journal is indexed on PubMed Central,
MedLine, CAS, SciSearch®, Current Contents®/Clinical Medicine,
Journal Citation Reports/Science Edition, EMBase, Scopus and the
Elsevier Bibliographic databases. The manuscript management system
is completely online and includes a very quick and fair peer-review
system, which is all easy to use. Visit http://www.dovepress.com/
testimonials.php to read real quotes from published authors.
International Journal of Nanomedicine 2015:10
submit your manuscript | www.dovepress.com
Dovepress
Dovepress
Dovepress
7412
Yin et al
46. Frei E. Albumin binding ligands and albumin conjugate uptake by
cancer cells. Diabetol Metab Syndr. 2011;3(1):11.
47. Gao L, Liu G, Ma J, et al. Paclitaxel nanosuspension coated with P-gp
inhibitory surfactants: II. Ability to reverse the drug-resistance of
H460 human lung cancer cells. Colloid Surf B Biointerfaces. 2014;117:
122–127.
48. Fang J, Liao L, Yin H, Nakamura H, Shin T, Maeda H. Enhanced bacte-
rial tumor delivery by modulating the EPR effect and therapeutic poten-
tial of Lactobacillus casei. J Pharm Sci. 2014;103(10):3235–3243.
49. Lu J, Huang Y, Zhao W, et al. Design and characterization of PEG-
derivatized vitamin E as a nanomicellar formulation for delivery of
paclitaxel. Mol Pharm. 2013;10(8):2880–2890.
50. Gligorov J, Lotz JP. Preclinical pharmacology of the taxanes: implica-
tions of the differences. Oncologist. 2004;9(Suppl 2):3–8.
51. Gong J, Huo M, Zhou J, et al. Synthesis, characterization, drug-loading
capacity and safety of novel octyl modified serum albumin micelles.
Int J Pharm. 2009;376(1–2):161–168.
52. ten Tije AJ, Verweij J, Loos WJ. Sparreboom A. Pharmacological
effects of formulation vehicles: implications for cancer chemotherapy.
Clin Pharmacokinet. 2003;42(7):665–685.
... PTX liposome-albumin composite nanoparticles were designed to avoid the direct contact of albumin with toxic organic solvents and enhance the colloidal stability of the formulation (19). PTX-PEG-HSA complex was also developed by dispersing PTX into PEG scaffold without using organic OPEN ACCESS solvents or Cremophor EL as solubilizers and then formulated with HSA into nanosuspensions suitable for intravenous administration (25). Furthermore, the bovine serum albumin (BSA) nanogel was synthesized through reaction between amino groups and polymers in phosphate buffer at room temperature without any organic molecules (26), while ionic liquid-based method was also used as an alternative technique (27). ...
... The QTY code is a very simple and versatile protein design tool, which is based on 1.5 Å electron density maps of similar amino acids, for example, L:N/Q; V/I:T; F:Y (31,32). The QTY code has been successfully used to design several types of integral membrane proteins including GPCRs (25,26), cytokine receptors (60), glucose transporters (61), solute carrier transporters (62), by pairwise residue substitutions in the transmembrane region because nature has already evolved 3 distinct chemical types of alpha-helix including I) hydrophilic, II) hydrophobic, and III) amphiphilic types (63). It is believed that QTY code is reversibly applicable due to their unique structural correlation between polar and nonpolar amino acids. ...
Article
Full-text available
Human serum albumin (HSA) is a highly water-soluble protein with 67% alpha-helix content and three distinct domains (I, II, and III). HSA offers a great promise in drug delivery with enhanced permeability and retention effect. But it is hindered by protein denaturation during drug entrapment or conjugation that result in distinct cellular transport pathways and reduction of biological activities. Here we report using a protein design approach named reverse-QTY (rQTY) code to convert specific hydrophilic alpha-helices to hydrophobic to alpha-helices. The designed HSA undergo self-assembly of well-ordered nanoparticles with highly biological actives. The hydrophilic amino acids, asparagine (N), glutamine (Q), threonine (T), and tyrosine (Y) in the helical B-subdomains of HSA were systematically replaced by hydrophobic leucine (L), valine (V), and phenylalanine (F). HSArQTY nanoparticles exhibited efficient cellular internalization through the cell membrane albumin binding protein GP60, or SPARC (secreted protein, acidic and rich in cysteine)-mediated pathways. The designed HSArQTY variants displayed superior biological activities including: i) encapsulation of drug doxorubicin, ii) receptor-mediated cellular transport, iii) tumor cell targeting, and iv) antitumor efficiency compare to denatured HSA nanoparticles. HSArQTY nanoparticles provided superior tumor targeting and antitumor therapeutic effects compared to the albumin nanoparticles fabricated by antisolvent precipitation method. We believe that the rQTY code is a robust platform for specific hydrophobic modification of functional hydrophilic proteins with clear-defined binding interfaces.
... Among successful drug nanocarriers that are now established, albumin-bound NPs have opened up an avenue as an effective nanocarrier in cancer therapeutics [19]. Albumin can closely and reversibly bind to hydrophobic paclitaxel through noncovalent bonds to enable in vivo transport and release of transported substances and has become a natural carrier of hydrophobic substances [20][21][22]. It can also provide improved efficacy and tolerability compared to cremophor-based paclitaxel solutions [23,24]. ...
... The first emulsion was obtained followed by high-pressure homogenization (NanoLyzer-N2, Cogene, Hsinchu, Taiwan). A homogenization pressure (15,000 and 20,000 psi) was applied to the emulsion, and the number of homogenization cycles (10,20,30, and 40 cycles) was also optimized. The emulsion was subjected to various homogenization cycles, then passed through the homogenizer valve, and collected through a connecting tube at the base of the assembly, thus forming nano-sized emulsion droplets. ...
Article
Full-text available
Histone deacetylase (HDAC) inhibitors have emerged as a new class of antitumor agent for various types of tumors. MPT0B291, a novel selective inhibitor of HDAC6, demonstrated significant antiproliferative activity in various human cancer cell types. However, MPT0B291 has very low water solubility, which limits its clinical use for cancer therapy. In the current study, MPT0B291 was encapsulated in human serum albumin (HSA), and its anticancer activities were investigated. Nanoparticles (NPs) were prepared using two-stage emulsification resulting in 100~200-nm NPs with a fine size distribution (polydispersity index of <0.3). The in vitro drug release profiles of MPT0B291-loaded HSA NPs presented sustained-release properties. The cytotoxic effect on MIA PaCa-2 human pancreatic carcinoma cells was found to be similar to MPT0B291-loaded HSA NPs and the free-drug group. The albumin-based formulation provided a higher maximum tolerated dose than that of a drug solution with reduced toxicity toward normal cells. Furthermore, in vivo pharmacokinetic studies demonstrated an effective increase (5~8-fold) in the bioavailability of NPs containing MPT0B291 loaded in HSA compared to the free-drug solution with an extended circulation time (t1/2) leading to significantly enhanced efficacy of anticancer treatment.
... Abraxane™ is the first FDA-approved chemotherapeutic formulation based on Nab™ nanotechnology, which relies heavily on the use of organic solvents, namely, chloroform. 25 The toxicity introduced by residual chloroform poses a potential risk to patient health. In response to the issue of chronic toxicity, a reversible self-assembling method, which eliminates the dependence on toxic organic solvents during manufacturing, was developed in a preliminary study and demonstrated to be capable of successfully preparing HSAbound PTX and CFZ nanosuspensions. ...
... Furthermore, both NPs formed using this method still retained their suitability for intravenous (IV) administration. 25 Therefore, in this study, the preparation of CFZ-loaded, PTX-loaded, and CFZ/PTX co-loaded HSA NPs was developed and optimized. To confirm these advantages, the properties of the three drug-loaded HSA NPs, including the encapsulating efficiency (EE), drug-loading (DL), mean size, polydispersity index (PDI), drug release, and cell growth inhibition against MIA CaPa-2 cells (human pancreatic cancer cell line) were characterized in vitro. ...
Article
Full-text available
Purpose: Therapeutic efficacy of pancreatic adenocarcinomas (PACs) with combined therapy of carfilzomib (CFZ) and paclitaxel (PTX) co-loaded in human serum albumin (HSA) nanoparticles (NPs) was examined. Methods: CFZ and PTX were encapsulated individually or combined into HSA NPs by a simple reverse self-assembly method developed to achieve an optimal combination ratio for synergistic therapy. CFZ or/and PTX loaded HSA nanoparticles were physically characterized and the evaluation of combination index, drug release, pharmacokinetic, anti-tumor, and biodistribution studies were conducted. Results: All resultant drug-loaded HSA NPs were spherical with a particle size of <150 nm and a zeta potential of -21.1~-23.0 mV. Drug loading rates and entrapment efficiencies were 9.1%~10.1% and 90.7%~97.1%, respectively. CFZ and PTX demonstrated synergistic effects in an MIA PaCa-2 cytotoxicity at a 1:2 ratio (CI50 were 0.01~0.25). In vitro dissolution revealed that the CFZ/PTX ratio released from the co-loaded HSA NPs (CFZ/PTX/HSA NPs) was about 1.77~2.08, which conformed to the designated loaded ratio. In vivo evaluation showed that the combined therapy of CFZ and PTX at a 1:2 ratio co-loaded in HSA NPs (CFZ/PTX/HSA NPs) demonstrated optimal synergistic improvement of the growth inhibition of MIA PaCa-2 cells with less systematic toxicity, even though the pharmacokinetic profiles observed did not show obvious beneficial and their biodistributions in tumors were found to be smaller. Conclusion: The one-pot reverse assembly method developed was environmentally friendly and capable of co-loading an optimal combination ratio of two chemodrugs into HSA NPs for synergistic therapy.
... Synthetic and semisynthetic NPs have revolutionized medicine and need to use a substantial amount of organic toxic materials and solvents. Despite intriguing benefits (targeted drug delivery, high drug solubility, high drug accumulation, improved bioavailability, and increased systemic circulation stability) of these synthetic NPs, complete removal of organic solvents (chloroform and cremophor-EL) is still challenging and limited commercial translation for their clinical use exist Yin et al., 2015). Thus, scientists have opted for an alternative future-focused biogenic NP synthesis using fruit/vegetable waste containing the aforementioned bioactive compounds. ...
Article
Nanotechnology is a continually growing field with a wide range of applications from food science to biotechnology and nanobiotechnology. As the current world is grappling with non-biodegradable waste, considered more challenging and expensive to dispose of than biodegradable waste, new technologies are needed today more than ever. Modern technologies, especially nanotechnology, can transform biodegradable waste into products for human use. Researchers are exploring sustainable pathways for nanotechnology by utilizing biodegradable waste as a source for preparing nanomaterials. Over the past ten years, the biogenic production of metallic nanoparticles (NPs) has become a promising alternative technique to traditional NPs synthesis due to its simplicity, eco-friendliness, and biocompatibility in nature. Fruit and vegetable waste (after industrial processing) contain various bioactives (such as flavonoids, phenols, tannins, steroids, triterpenoids, glycosides, anthocyanins, carotenoids, ellagitannins, vitamin C, and essential oils) serving as reducing and capping agents for NP synthesis and they possess antibacterial, antioxidant, and anti-inflammatory properties. This review addresses various sources of biogenic NPs including their synthesis using fruit/vegetables waste, types of biogenic NPs, extraction processes and extracted biomaterials, the pharmacological functionality of NPs, industrial aspects, and future perspectives. In this manner, this review will cover the most recent research on the biogenic synthesis of NPs from fruit/vegetable peels to transform them into therapeutic nanomedicine.
... One way to improve the apparent aqueous solubility of such drugs is via their dissolution in water miscible, polar organic solvents or their solubilisation within surfactant micelles [3]. Unfortunately, the intravenous administration of hydrophobic drugs in polar organic solvents frequently results in toxic side effects [4][5][6][7]. In contrast, if the same drug molecules are incorporated in micelles, the need for potentially harmful solvents and the pain associated with their intravenous administration can be avoided or, at least, minimised [2,8]. ...
Article
Full-text available
The solubilisation capacities of the micelles formed by a range of surfactants possessing a dodecyl (C12) hydrocarbon ‘tail’ and a variety of hydrophilic ‘headgroups’ have been studied for a range of hydrophobic steroids (testosterone (T) and its propionate (TP) and enanthate (TE) esters). The solubilisation studies were performed at ambient temperature, with drug concentration monitored (over 48-72 hours) by UV absorbance. The various surfactant systems were characterised in terms of their physicochemical properties, including their surface tension, critical micelle concentration, viscosity, and density. Results show that while the apparent aqueous solubility of the poorly water-soluble steroids increases upon solubilisation within the surfactant micelles, the extent of this increase varies with the nature of the hydrophilic headgroup and the hydrophobicity of the drug. Specifically, changing the surfactant headgroup from non-ionic to zwitterionic, then to ionic, results in a marked increase in solubility of the two least hydrophobic steroids, testosterone and testosterone propionate, suggesting rather counter-intuitively that these steroidal drugs tend to associate to a greater extent with micelles formed by surfactants with more hydrophilic/charged head groups. The more hydrophobic steroid, TE, was solubilised to the greatest extent, being predominantly solubilised within the micelle core. In this case, the influence of surfactant head group on TE solubilisation was less obvious, with surfactants containing ionic and zwitterionic head groups exhibiting high levels of solubilisation. The physicochemical properties that had most influence on solubilisation of the testosterone analogues, as evaluated using multiple linear regression, were micelle shape for T and TP, and critical micelle concentration for TE. Through the findings of this comparative study, the essential molecular descriptors of optimal solubilisation for improved drug delivery and the loci of solubilisation of the testosterone analogues have been identified.
... Ptx, a natural isoprenoid derivative with potent anticancer activity stabilizes polymerized microtubules causing defects in mitotic spindle assembly, chromosome segregation, and cell division [1][2][3][4][5]. Ptx is widely used to perform chemotherapy treatments in cancer patients [3,4,6,7], however, its toxicity, resistance, and low water solubility limit its therapeutic efficacy [8][9][10]. In order to reduce these obstacles and achieve less toxic and more effective parenteral administration of taxanes such as Ptx, different excipients and vehicles are being extensively investigated [1,7,[11][12][13]. ...
Article
Full-text available
The use of nanoparticles is one of the strategies currently studied to minimize the toxicity and lack of tissue specificity of many cancer drugs used in chemotherapy. In this research the physicochemical and biological behavior of a novel self-assembled nanostructure of the antibiotic Teicoplanin (Teico) was characterized as a nanocarrier system for solubilizing highly hydrophobic drugs like Paclitaxel (Ptx) in aqueous media. The Teico micelles were loaded with Ptx in DMSO or PEG-400. The interaction between the loaded micelles and Albumin (HSA) was then studied by size exclusion chromatography. Transmission electron microscopy, dynamic light scattering and high-resolution liquid chromatography were also used to characterize the physicochemical and structural properties of the micelles to form the Teico/Ptx and Teico/Ptx /HSA micelles. Cellular uptake of Ptx was evaluated by fluorescent microscopy. The in vitro cytotoxicity of the complexes was studied on Hep-2 tumor cells, by a Crystal Violet assay. Teico cosolvent-free micelles can solubilize up to 20 mg.mL-1 of Ptx dissolved in PEG, increasing four times the solubility of Ptx in water compared to Abraxane, and 20,000 times the intrinsic solubility of Ptx in water. In addition, Teico/Ptx micelles binds spontaneously HSA through hydrophobic interaction. Teico and Teico/HSA micelles as a Ptx transporter does not affect its release or biological activity. Therefore, Teico/Ptx or Teico/Ptx/HSA complexes appear as new alternatives for transporting larger amounts of hydrophobic drugs that offer advantages, turning it an interesting option for further study.
... Albumin can bind closely and reversibly with hydrophobic PTX through noncovalent bonds to realize the transport and release of the transported substances in vivo and has become a natural carrier of hydrophobic substances (Yin et al., 2015;Thadakapally et al., 2016;Tai et al., 2017). In the present study, we used albumin to prepare Nab-PTX-PA via Nab TM technique. ...
Article
Full-text available
Background Combination of the prodrug technique with an albumin nano drug-loaded system is a novel promising approach for cancer treatment. However, the long-lasting and far-reaching challenge for the treatment of cancers lies in how to construct the albumin nanometer drug delivery system with lead compounds and their derivatives. Methods In this study, we reported the preparation of injectable albumin nanoparticles (NPs) with a high and quantitative drug loading system based on the NabTM technology of paclitaxel palmitate (PTX-PA). Results Our experimental study on drug tissue distribution in vivo demonstrated that the paclitaxel palmitate albumin nanoparticles (Nab-PTX-PA) remained in the tumor for a longer time post-injection. Compared with saline and paclitaxel albumin nanoparticles (Abraxane®), intravenous injection of Nab-PTX-PA not only reduced the toxicity of the drug in normal organs, and increased the body weight of the animals but maintained sustained release of paclitaxel (PTX) in the tumor, thereby displaying an excellent antitumor activity. Blood routine analysis showed that Nab-PTX-PA had fewer adverse effects or less toxicity to the normal organs, and it inhibited tumor cell proliferation more effectively as compared with commercial paclitaxel albumin nanoparticles. Conclusions This carrier strategy for small molecule drugs is based on naturally evolved interactions between long-chain fatty acids (LCFAs) and Human Serum Albumin (HSA), demonstrated here for PTX. Nab-PTX-PA shows higher antitumor efficacy in vivo in breast cancer models. On the whole, this novel injectable Nab-PTX-PA has great potential as an effective drug delivery system in the treatment of breast cancer.
Article
Introduction: Intravenous nanocrystals (INCs) have shown intrinsic advantages in antitumor applications, particularly their properties of high drug loading, low toxicity, and controllable size. Therefore, it has a very bright application prospect as a drug delivery system. Areas covered: The ideal formulation design principles, fabrication, solidification, in vivo fate of INCs, the applications in drug delivery system (DDS) and the novel applications are covered in this review. Expert opinion: It is vital to select a suitable formulation and fabrication method to produce a stable and sterile INCs. Besides, the type of stabilizers and physical characteristics can also influence the in vivo fate of INCs, which is worthy of further studying. Based on wide researches about applications of INCs in cancer, biomimetic INCs are concerned increasingly for its favorable compatibility. The output of these studies suggested that INCs-based drug delivery could be a novel strategy for addressing the delivery of the drug that faces solubility, bioavailability, and toxicity problems.
Article
The poor aqueous solubility is a well-recognized restriction for the clinical application of many drug molecules. Micelles delivery system provides a promising strategy for the solubility enhancement of hydrophobic drugs. This study developed and evaluated different polymeric mixed micelles prepared using hot-melt extrusion coupled with the hydration method to improve the solubility and extend the release of the model drug ibuprofen (IBP). The physicochemical properties of the prepared formulations were characterized in terms of particle size and zeta potential, surface morphology, crystallinity, encapsulation efficiency, drug content, in vitro drug release, dilution stability, and storage stability. Soluplus®/poloxamer 407, Soluplus®/poloxamer 188, and Soluplus®/TPGS mixed micelles had average particle sizes of 86.2 ± 2.8, 89.6 ± 4.2, and 102.5 ± 3.13 nm, respectively with adequate encapsulation efficiencies of 80% to 92%. Differential scanning calorimetry (DSC) studies confirmed that the IBP molecules were dissolved in the polymers in an amorphous state. The in vitro release results revealed that the IBP-loaded mixed micelles presented extended-release behavior compared to the free drug. In addition, the developed polymeric mixed micelles remained stable upon dilution and one-month storage. These results demonstrated that the hot-melt extrusion coupling hydration method could be a promising, effective, and environment-friendly manufacturing technique for the scale-up production of polymeric mixed micelles to deliver insoluble drugs.
Article
The objective of this study was to develop novel herpetrione (HPE) nanosuspensions stabilized by glycyrrhizin (HPE-NSs/GL) for enhancing bioavailability and hepatoprotective effect of HPE. HPE-NSs/GL were prepared by wet media milling method and then systemically evaluated by particle size analysis, scanning electronic microscopy (SEM), X-ray powder diffraction (XRPD), dissolution test, pharmacokinetics, and hepatoprotective effect. HPE-NSs stabilized by poloxamer 407 (HPE-NSs/P407) were also prepared and used as reference for comparison. HPE-NSs/GL and HPE-NSs/P407 with similar particle size around 450 nm and PDI less than 0.2 were successfully prepared and both of them appeared to be spherical under SEM. The XRPD results demonstrated that HPE in both HPE NSs/GL and HPE NSs/P407 was presented in amorphous state and the addition of GL or P407 and milling process didn't alter the physical state of HPE. The dissolution and pharmacokinetics studies demonstrated that HPE NSs/GL exhibited significant enhancement in drug dissolution (72.44% within 24 h) and AUC0-t (24.91 ± 3.3 mg/L·h) as compared to HPE coarse suspensions (HPE CS, 34.19% & 13.07 ± 1.02 mg/L·h), but was similar with those of HPE-NSs/P407 (80.06% & 26.75 ± 4.06 mg/L•h). Moreover, HPE NSs/GL exhibited significantly better hepatoprotective effect as compared to HPE CS and HPE NSs/P407 as indicated by the lowering of the elevated serum ALT and AST levels and the improvement of the hepatic morphology and architecture, which might be attribute to the improved bioavailability of HPE, and synergistic hepatoprotective effect of GL via alleviating inflammation evidenced by the significant decreased hepatic levels of inflammatory cytokines IL-1β, IL-6 and TNF-α. It could be concluded that GL might be an effective stabilizer for preparing HPE NSs, and HPE-NSs/GL is a potential formulation strategy for improving oral bioavailability and hepatoprotective effect of HPE.
Article
Full-text available
The non-ionic surfactants Cremophor® EL (CrEL; polyoxyethyleneglycerol triricinoleate 35) and polysorbate 80 (Tween® 80; polyoxy-ethylene-sorbitan-20-monooleate) are widely used as drug formulation vehicles, including for the taxane anticancer agents paclitaxel and docetaxel. A wealth of recent experimental data has indicated that both solubilisers are biologically and pharmacologically active compounds, and their use as drug formulation vehicles has been implicated in clinically important adverse effects, including acute hypersensitivity reactions and peripheral neuropathy. CrEL and Tween® 80 have also been demonstrated to influence the disposition of solubilised drugs that are administered intravenously. The overall resulting effect is a highly increased systemic drug exposure and a simultaneously decreased clearance, leading to alteration in the pharmacodynamic characteristics of the solubilised drug. Kinetic experiments revealed that this effect is primarily caused by reduced cellular uptake of the drug from large spherical micellar-like structures with a highly hydrophobic interior, which act as the principal carrier of circulating drug. Within the central blood compartment, this results in a profound alteration of drug accumulation in erythrocytes, thereby reducing the free drug fraction available for cellular partitioning and influencing drug distribution as well as elimination routes. The existence of CrEL and Tween® 80 in blood as large polar micelles has also raised additional complexities in the case of combination chemotherapy regimens with taxanes, such that the disposition of several coadministered drugs, including anthracyclines and epipodophyllotoxins, is significantly altered. In contrast to the enhancing effects of Tween® 80, addition of CrEL to the formulation of oral drug preparations seems to result in significantly diminished drug uptake and reduced circulating concentrations. The drawbacks presented by the presence of CrEL or Tween® 80 in drug formulations have instigated extensive research to develop alternative delivery forms. Currently, several strategies are in progress to develop Tween® 80- and CrEL-free formulations of docetaxel and paclitaxel, which are based on pharmaceutical (e.g. albumin nanoparticles, emulsions and liposomes), chemical (e.g. polyglutamates, analogues and prodrugs), or biological (e.g. oral drug administration) strategies. These continued investigations should eventually lead to more rational and selective chemotherapeutic treatment.
Article
Full-text available
Learning Objectives After completing this course, the reader will be able to: Identify the differences in molecular pharmacology of the taxanes. Describe the impact of the pharmacokinetic and pharmacodynamic profiles of the taxanes on their administration and toxicity. Outline the toxicity profiles of paclitaxel and docetaxel. Access and take the CME test online and receive 1 hour of AMA PRA category 1 credit at CME.TheOncologist.com Taxanes are one of the most powerful classes of compounds among all chemotherapeutic drugs. Only 30 years separate the isolation of the first taxane from the results of direct clinical comparisons in metastatic breast, ovarian, and lung cancer between the two taxanes available in routine clinical practice. These results suggest a more favorable benefit-to-risk ratio for docetaxel compared to paclitaxel when these drugs are used as single agents or in combination with other chemotherapeutic agents in an every-3-week dosing regimen. Pharmacological data support the difference between the taxanes, likely explaining the clinical results. Considering the molecular pharmacology of the two drugs, docetaxel appears to bind to β-tubulin with greater affinity and has a wider cell cycle activity than paclitaxel. Docetaxel also appears to have direct antitumoral activity via an apoptotic effect mediated by bcl-2 phosphorylation. In addition, docetaxel has a longer retention time in tumor cells than paclitaxel because of greater uptake and slower efflux. Pharmacokinetics and pharmacodynamics of the taxanes show both agents to be extensively metabolized in the liver, and paclitaxel has a nonlinear pharmacokinetic behavior while docetaxel has linear pharmacokinetics. These differences explain the more simple treatment schedule and favorable results for docetaxel as a single agent and in combination therapy. Last, but not least, there is a pharmacokinetic interaction between paclitaxel and the anthracyclines, an active class of compounds commonly used in the treatment of breast cancer. This pharmacokinetic interaction is associated with greater cardio- and myelotoxicities, which are sequence dependent. These pharmacological data likely explain the different clinical development strategies for the two molecules as well as the different clinical results from individual trials and direct comparisons.
Article
Full-text available
Based on the clinical fact that paclitaxel (PTX) intravenous injection frequently causes hypersensitive reaction, its oral administration is vital for its clinical development. However, the development oral PTX formulation is difficult due to its low oral bioavailability caused by its low permeability, low solubility, efflux and affinity for intestinal and liver metabolic enzymes. In an effort to develop an alternative formulation of PTX suitable for oral administration, PTX-loaded nanocochleates (PTXNC) were developed by addition of calcium ion into preformed nanoliposomes (PTXNL) comprising PTX, phosphatidylcholine and cholesterol and evaluated by in vitro and in vivo methods in comparison with PTXNL and pure PTX. Stable tubular rod structure of PTXNC possessing particle size, encapsulation efficiency and zeta potential of 269 ± 2 nm, 71.12 ± 1.87% and −3.9 mV, respectively were obtained from homogenous unilamellar, discrete and spherical structured nanoliposomes with diameter and zeta potential of 247 ± 3 nm and −12.3 mV, respectively. Powder X-ray diffraction and thermal study revealed that PTX was partial crystalline and amorphous state in PTXNL and PTXNC, respectively. PTXNC demonstrated controlled release of PTX. PTXNC showed many fold improvement in vitro anticancer activity towards A-549 (human lung adenocarcinoma cells), ovarian cancer OVCAR-3 and breast cancer MCF-7 cells with a 14- and 19-fold increase in oral bioavailability in rats as compared to PTXNL and pure PTX, respectively. Moreover, PTXNC showed low tissue distribution. These results collectively suggest that nanocochleates could therefore advantageously be employed to improve anticancer activity of PTX and an alternative to the present intravenous administration.
Article
Paclitaxel (PTX) is a first line chemotherapy drug for breast cancer. There have been few studies reported concerning the therapeutic efficacy of paclitaxel-conjugated polymeric micelles in breast cancer in vivo. Two kinds of PTX conjugate micelles, one of which (M(PTX)) contained 25wt.% of PTX and the other (M(FA/PTX)) contained 22.5wt.% of PTX and 1.4wt.% of folate (FA), were prepared for cell apoptosis and anti-tumor activity evaluation on EMT-6 mice breast cancer models in comparison with 0.9wt.% saline (control) and equivalent PTX. Cell apoptosis was analyzed by flow cytometry. Breast tumors were examined histologically with H&E staining and immunohistochemically by examining Bax and Bcl-2 expression. The survival status of tumor-bearing mice with different treatments was also examined. On day 5 of the drug administration, the average tumor masses were 0.49, 0.33, 0.22, and 0.18g for the control, PTX, M(PTX) and M(FA/PTX) groups, respectively. The inhibition rates of tumor growth calculated for the three drug groups were 32.6%, 51.6% and 62.3%, respectively. The percentage of cell apoptosis based on flow cytometry was 1.0%, 36.6%, 55.9% and 66.1%, respectively, which showed statistically significant differences (p<0.05) between three drug groups and the control group. Bcl-2 expression of PTX and M(FA/PTX) groups was lower than control group (p<0.05). Bax expression of drug groups was higher than control group (p<0.05). At an equivalent paclitaxel dose of 26.7mg/kg, the average survival time was 33days, 31days, 34days and 42days, respectively (p<0.05). The M(FA/PTX) have better anti-tumor activity and are promising in treatment of human breast cancers. Copyright © 2015 Elsevier B.V. All rights reserved.
Article
Intravital imaging of nanoparticle extravasation and tumor accumulation has revealed, for the first time, detailed features of carrier and drug behavior in circulation and tissue that suggest new directions for optimization of drug nanocarriers. Using intravital fluorescent microscopy, the extent of the extravasation, diffusion in the tissue, internalization by tissue cells, and uptake by the RES system were studied for polymeric micelles, nanoemulsions, and nanoemulsion-encapsulated drug. Discrimination of vascular and tissue compartments in the processes of micelle and nanodroplet extravasation and tissue accumulation was possible. A simple 1-D continuum model was suggested that allowed discriminating between various kinetic regimes of nanocarrier (or released drug) internalization in tumors of various sizes and cell density. The extravasation and tumor cell internalization occurred much faster for polymeric micelles than for nanoemulsion droplets. Fast micelle internalization resulted in the formation of a perivascular fluorescent coating around blood vessels. A new mechanism of micelle extravasation and internalization was suggested, based on the fast extravasation and internalization rates of copolymer unimers while maintaining micelle/unimer equilibrium in the circulation. The data suggested that to be therapeutically effective, nanoparticles with high internalization rate should manifest fast diffusion in the tumor tissue in order to avoid generation of concentration gradients that induce drug resistance. However an extra-fast diffusion should be avoided as it may result in the flow of extravasated nanoparticles from the tumor to normal organs, which would compromise targeting efficiency. The extravasation kinetics were different for nanodroplets and nanodroplet-encapsulated drug F-PTX suggesting a premature release of some fraction of the drug from the carrier. In conclusion, the development of an “ideal” drug carrier should involve the optimization of both drug retention and carrier diffusion parameters.
Article
The aim of the study was to develop stable Paclitaxel nanocrystals (PTX/NCs) for enhanced oral delivery of Paclitaxel (PTX) by circumventing its difficult solubilization properties and rapid metabolism. Preparation of Nanocrystals (NCs) was carried out using high pressure homogenizer (Microfluidizer(TM)) without using any organic solvent. Effect of various process and formulation parameters on development and stability of nanocrystals (NCs) were investigated. Particle characteristics, stability studies, in-vitro cellular studies and oral pharmacokinetics in male Wistar rats were examined. It was found that different stabilizer used had different effect on size reduction and stability. Surfactants (Tween 80) and low molecular weight synthetic polymer Sodium poly styrene sulfonate (PSS) found more suitable and efficient compared to high molecular weight polymers glycol chitosan (GC) and sodium alginate (SA). In-vitro cytotoxicity and cell cycle arrest studies on MCF7 and MDA-MB breast cancer cell lines revealed that PTX/NCs retained the activity even after processing at high pressure and also NCs were more potent and efficacious than PTX solution. The oral in-vivo pharmacokinetic studies demonstrated that PTX/NCs exhibit significant increase in AUC0-t, Cmax, MRT and decrease in Tmax, compared to plain PTX crystals. The increase in AUC was almost 9-10 fold compared to plain PTX crystals. Altogether study showed that PTX/NC can be a clinically relevant drug delivery system for oral chemotherapy as it can remarkably increases the pharmacological effect by increasing oral bioavailability. Copyright © 2014. Published by Elsevier B.V.
Article
Nanoparticles are efficient delivery vehicles for cancer therapy such as paclitaxel (PTX). In this study, we formulated PTX into PLGA polymeric nanoparticles. Vitamin E TPGS was used as an emulsifier to stabilize the nanoparticle formulation. PTX was encapsulated in TPGS-emulsified polymeric nanoparticles (TENPs) by a nanoprecipitation method in ethanol-water system. The resultant PTX-TENPs showed a very uniform particle size (∼100nm) and high drug encapsulation (>80%). The cytotoxicity of PTX-TENPs was examined in A549 lung cancer cell line. Preferential tumor accumulation of TENPs was observed in the A549 lung cancer xenograft model. Tumor growth was significantly inhibited by intravenous injection of PTX-TENPs. Our results suggested that the modified nanoprecipitation method holds great potential for the fabrication of the PTX loaded polymeric nanoparticles. TPGS can be used in the manufacture of polymeric nanoparticles for the controlled release of PTX and other anti-cancer drugs. Copyright © 2014 Elsevier B.V. All rights reserved.