You are on page 1of 9

Colloids and Surfaces B: Biointerfaces 146 (2016) 152–160

Contents lists available at ScienceDirect

Colloids and Surfaces B: Biointerfaces


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

Cationic drug-based self-assembled polyelectrolyte complex micelles:


Physicochemical, pharmacokinetic, and anticancer activity analysis
Thiruganesh Ramasamy a , Bijay Kumar Poudel a , Himabindu Ruttala a , Ju Yeon Choi a ,
Truong Duy Hieu a , Kandasamy Umadevi b , Yu Seok Youn c , Han-Gon Choi d ,
Chul Soon Yong a,∗ , Jong Oh Kim a,∗
a
College of Pharmacy, Yeungnam University, 214-1 Dae-dong, Gyeongsan 712-749, South Korea
b
St. Paul’s College of Pharmacy, Osmania University, Hyderabad, Telangana, India
c
School of Pharmacy, SungKyunKwan University, 300 Cheoncheon-dong, Jangan-gu, Suwon, 440-746, South Korea
d
College of Pharmacy, Institute of Pharmaceutical Science and Technology, Hanyang University, 55, Hanyangdaehak-ro, Sangnok-gu, Ansan 426-791,
South Korea

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

Article history: Nanofabrication of polymeric micelles through self-assembly of an ionic block copolymer and oppositely
Received 22 March 2016 charged small molecules has recently emerged as a promising method of formulating delivery systems.
Received in revised form 31 May 2016 The present study therefore aimed to investigate the interaction of cationic drugs doxorubicin (DOX)
Accepted 2 June 2016
and mitoxantrone (MTX) with the anionic block polymer poly(ethylene oxide)-block-poly(acrylic acid)
Available online 5 June 2016
(PEO-b-PAA) and to study the influence of these interactions on the pharmacokinetic stability and anti-
tumor potential of the formulated micelles in clinically relevant animal models. To this end, individual
Keywords:
DOX and MTX-loaded polyelectrolyte complex micelles (PCM) were prepared, and their physicochemical
Nanofabrication
Polyelectrolyte complex micelles properties and pH-responsive release profiles were studied. MTX-PCM and DOX-PCM exhibited a differ-
Cationic drugs ent release profile under all pH conditions tested. MTX-PCM exhibited a monophasic release profile with
Pharmacokinetic no initial burst, while DOX-PCM exhibited a biphasic release. DOX-PCM showed a higher cellular uptake
Anticancer activity than that shown by MTX-PCM in A-549 cancer cells. Furthermore, DOX-PCM induced higher apoptosis
of cancer cells than that induced by MTX-PCM. Importantly, both MTX-PCM and DOX-PCM showed pro-
longed blood circulation. MTX-PCM improved the AUCall of MTX 4-fold compared to a 3-fold increase
by DOX-PCM for DOX. While a definite difference in blood circulation was observed between MTX-PCM
and DOX-PCM in the pharmacokinetic study, both MTX-PCM and DOX-PCM suppressed tumor growth
to the same level as the respective free drugs, indicating the potential of PEGylated polymeric micelles
as effective delivery systems. Taken together, our results show that the nature of interactions of cationic
drugs with the polyionic copolymer can have a tremendous influence on the biological performance of a
delivery system.
© 2016 Elsevier B.V. All rights reserved.

1. Introduction tic drugs, various drug delivery systems have been developed.
Among them, block copolymer-based self-assembled polymeric
Conventional chemotherapeutic approach is the main treat- micelles have demonstrated promising potential in the delivery
ment option for cancer [1]. Despite great strides made in of anticancer drugs. The nanosized micelles offer many advan-
understanding cancer biology, conventional chemotherapeutic tages, including uniform size distribution, core-shell architecture,
drugs are characterized by non-specific distribution and high accu- high drug loading, and physical stability [3,4]. Polyethylene glycol
mulation in healthy cells, leading to dose-limiting side effects (PEG) is widely used to graft the hydrophobic part of amphiphilic
that seriously impede their clinical application [2]. To minimize polymers and form the outer shell of the micelles. Such polymeric
side effects and improve therapeutic efficacy of chemotherapeu- micelles have been shown to increase the systemic circulation time
of drugs and preferentially accumulate in tumors via enhanced
permeability and retention (EPR) effect [5].
Polyelectrolyte complex micelles (PCM), a special class of
∗ Corresponding authors. micelles formed by electrostatic interaction of opposite charged
E-mail addresses: csyong@ynu.ac.kr (C.S. Yong), jongohkim@yu.ac.kr (J.O. Kim).

http://dx.doi.org/10.1016/j.colsurfb.2016.06.004
0927-7765/© 2016 Elsevier B.V. All rights reserved.
T. Ramasamy et al. / Colloids and Surfaces B: Biointerfaces 146 (2016) 152–160 153

species (ionic blocks), have recently been developed [6,7]. For drug of the polymer (PEO-b-PAA) and drug (MTX or DOX) were pre-
delivery applications, therapeutic moieties (small molecules, DNA, pared separately and mixed together at various charge ratios
or proteins) act as a charged species in the formation of PCM of amino groups of drugs to carboxylate groups of the polymer
[8]. As a result, PCM self-assemble into a nanoscale core-corona (R = [drug]/[COO− ]). The mixture was incubated at 25 ◦ C for 24 h
structure with ionic segment-drug complex as the core and the during which the drug and the polymer block self-assembled to
water-soluble nonionic segments (PEG) as the outer corona. The form core-shell micelles. The pH of the solution mixture was
neutral polymer segment, which forms the corona of the PCM, changed to investigate the effect of pH on the degree of ioniza-
ensures aqueous solubility under stoichiometric conditions. Fur- tion and micelle forming. The free unbound drugs were removed
thermore, the hydrophilic shell prevents the aggregation and phase thoroughly by repeated filtrations using Amicon YM-10 centrifu-
separation of micelles, and improves their stability [9]. gal filter devices (MWCO, 10000 Da; Millipore, Billerica, MA, USA)
It is well known that stability of electrostatically assembled PCM pretreated with drugs to retain only the drug-loaded micelles. The
relies on the nature and charge density of ionic species involved. concentration of MTX or DOX in the filtrate was estimated by
The nature of the interaction determines its binding strength. The UV/VIS spectrophotometry (Perkin Elmer U-2800, Hitachi, Japan).
stronger the charge interaction between the ionic segments, the The wavelengths of 609 and 485 nm were selected for measuring
stronger and more stable will the micellar complex be, which lays MTX and DOX, respectively.
the foundation for its in vivo stability. Obtaining in vivo stability
represents a challenge for the successful delivery of nanoparticles
2.3. Particle size and ␨-potential analysis
to tumor interstitial spaces [10].
Well-known anticancer drugs doxorubicin (DOX) and mitox-
Particle size (nm), polydispersity index (PDI), and zeta (␨)-
antrone (MTX) are anthracyclines with a broad spectrum of activity
potential (mV) of MTX-PCM and DOX-PCM were analyzed by
against a variety of cancers including breast, lung, prostate, bone,
dynamic light scattering (DLS). Zetasizer Nano ZS (Malvern Instru-
and bladder cancers. These anticancer agents act by intercalating
ments, Malvern, UK) equipped with He–Ne laser was used to
DNA and inhibiting topoisomerase II [11]. While both DOX and MTX
measure the particle size. A fixed angle of 90◦ was selected and the
are anthracycline moieties, they differ in number and substitution
laser was operated at 635 nm. Nano DTS software (version 6.34)
states of amino functionalities present. DOX consists of four fused
was employed to analyze the size, PDI, and surface charge of the
rings with a sugar moiety containing a primary amine group (-NH2 ),
micelles. Each measurement was performed in triplicate.
while MTX has three fused rings containing two secondary amine
groups (-NH-) [6,11]. Both DOX and MTX are positively charged at
physiological pH with an average pKa of ∼ 8, which is responsible 2.4. Morphological analysis
for electrostatic interactions with the carboxylate group (pKa ∼ 5)
of the block copolymer [12]. In the present study, poly(ethylene Transmission electron microscopy (TEM) (CM 200 UT; Philips,
oxide)-block-poly(acrylic acid) (PEO-b-PAA) was used as the block Andover, MA, USA) was used to characterize the morphology of
copolymer. The PCM were formed by the electrostatic interaction of drug-loaded PCM. The particles were observed at an accelerating
the protonated amino groups of MTX or DOX with the carboxylate voltage of 100 kV. Briefly, a drop of micellar dispersion (R = 0.5) was
moiety of the PAA segment of the PEO-b-PAA polymer. placed in the carbon-coated copper grid and allowed to settle for
The present study aimed to investigate the interactions of differ- 10 min. Excess liquid was soaked out with tissue paper. The thin
ent cationic drugs with the anionic block polymer PEO-b-PAA and layer of particles was counter-stained by 2% phosphotungstic acid
to study the influence of these interactions on the pharmacokinetic (PTA) as a negative staining. The particles were subjected to infrared
stability and antitumor potential of the formed PCM in clinically radiation for 5 min.
relevant animal models. Towards this purpose, pH-responsiveness
and release profiles of individual drugs from drug-loaded PCM were
2.5. Physical state characterization
monitored. In addition, an in vivo pharmacokinetic study of PCM
(DOX-PCM and MTX-PCM) in rats and an antitumor efficacy study
The X-ray diffraction (XRD) patterns of free DOX, MTX, DOX-
in A-549 cancer cell-xenografted mouse models were performed.
PCM, and MTX-PCM were recorded using a vertical goniometer and
The effect of amine-functionalized anticancer drugs on the physic-
X-ray diffractometer (X’Pert PRO MPD diffractometer, Almelo, The
ochemical and biological responses of micellar nanocarriers was
Netherlands) to measure Ni-filtered CuK␣ radiation (voltage, 40 kV;
demonstrated.
current, 30 mA) scattered in the crystalline regions of the sample.
The patterns were recorded at a scanning rate of 5◦ /min over the
2. Materials and methods 10–60◦ diffraction angle (2 ␪) range at an ambient temperature.

2.1. Materials
2.6. In vitro release studies

Doxorubicin hydrochloride was supplied by Dong-A Phar-


The release profiles of drugs from MTX-PCM or DOX-PCM
maceutical Company (Yongin, South Korea). Mitoxantrone dihy-
were evaluated by dialysis. Phosphate-buffered saline (PBS, pH 7.4,
drochloride was purchased from Shaanxi Top Pharm Chemical
0.14 M NaCl) and acetate-buffered saline (pH 5.0, 0.14 M NaCl) were
Co. Ltd (Xi’an, China). Poly(ethylene oxide)-block-poly(acrylic acid)
used to simulate the physiological and tumor pH. In brief, 1 ml of
(PEO-b-PAA, MWs of PEO and PAA blocks were 5000 and 6800 Da,
micellar dispersion (1 mg equivalent of MTX and DOX at R = 0.5)
respectively) was procured from Polymer Source, Inc. (Quebec,
was sealed in membrane tubing (Spectra/Por® ; 3500 Da cutoff) and
Canada). All other chemicals were of reagent grade purity and were
placed at 37 ◦ C at 100 rpm. The samples were withdrawn at prede-
used without any further modifications.
termined times and replaced with equal amounts of fresh medium.
The samples were collected, filtered, and analyzed using UV–vis
2.2. Preparation of drug-loaded PCM spectrophotometry at 609 and 485 nm for MTX and DOX, respec-
tively. The amount of drug released was plotted against time. The
The MTX and DOX-loaded PCM were formed by a simple mixing release kinetics was analyzed by fitting the data to appropriate
method as we reported previously [6]. Briefly, aqueous solutions mathematical models.
154 T. Ramasamy et al. / Colloids and Surfaces B: Biointerfaces 146 (2016) 152–160

Fig. 1. Schemes illustrating preparation of drug-loaded PCM.

2.7. Cell culture Korea. The rats were divided into four groups with 4 rats in each
group.
A-549 small lung cancer cells were grown in RPMI 1640 medium
supplemented with 10% (v/v) fetal bovine serum (FBS) in the pres-
2.11. Administration and blood collection
ence of penicillin and streptomycin (100 U/mL and 0.1 mg/mL,
respectively). The cells were maintained under ambient conditions
The rats were held in a supine position. The right femoral artery
(37 ◦ C containing 5% CO2 ) in a T-75 flask and periodically sub-
was cannulated to collect the blood samples, while the left femoral
cultured.
artery was cannulated to administer the individual DOX and MTX
formulations as a single dose (5 mg/kg). 300 ␮L of prepared PCM
2.8. Cellular uptake analysis formulations were administered to each rat via a tail vein injec-
tion. The micelles formulated at a feeding ratio of R = 0.5 were
The cellular uptake of free drugs and drug-loaded PCM was employed. Blood samples (200 ␮L) were collected at designated
investigated in A-549 cancer cells using fluorescence-assisted cell intervals (0.25, 0.5, 1, 2, 4, 6, 8, 10, 12, and 24 h). The surgical open-
sorting (FACS). Briefly, cells were seeded at a density of 3 × 105 ings were immediately sealed with surgical sutures to ease pain and
cells/well in a 6-well plate and incubated overnight. The cells were increase the length of the study period. After blood was withdrawn,
treated with free DOX, free MTX, DOX-PCM, and MTX-PCM (in it was immediately centrifuged (Eppendorf, Hauppauge, NY, USA)
equivalent concentrations of 10 ␮g/mL) and incubated for the indi- at 13 000 rpm for 10 min so that plasma could be separated and
cated periods. The cells were washed twice with PBS and harvested. extracted for further analysis.
The cells were resuspended in 1 mL of PBS and analyzed in a flow
cytometer (FACSCalibur, BD Biosciences, San Jose, CA, USA).
2.12. Preparation and evaluation of plasma samples by HPLC

2.9. Apoptosis analysis 150 ␮L of plasma was mixed with 150 ␮L of methanol and
vortex-mixed for 30 min. The mixture was centrifuged at high
The cells were seeded at a density of 3 × 105 cells/well in a 6- speed; supernatant was separated and subjected to vacuum evap-
well plate and incubated overnight. The cells were treated with oration. The evaporated residue was reconstituted with mobile
free DOX, free MTX, DOX-PCM, and MTX-PCM (in equivalent con- phase and injected into the HPLC column (20 ␮L). Two differ-
centrations of 5 ␮g/mL) and incubated for 24 h. Next day, cells were ent mobile phases were used: sodium formate (80 nM)/methanol
washed, trypsinized, harvested, and washed again with cold PBS. (80/20; pH 2.9) for MTX and methanol/water/acetic acid (50/49/1;
The pellet was treated with 2.5 ␮L of Annexin V-FITC and 2.5 ␮L of pH 3) for DOX. Flow rate of the mobile phase was 1 ml/min and
7-AAD for 15 min at room temperature. The percentage of apop- effluents were measured at 254 and 480 nm for MTX and DOX,
totic cells was analyzed using a flow cytometer (FACSCalibur, BD respectively.
Biosciences, San Jose, CA, USA).

2.13. Pharmacokinetic parameters


2.10. Pharmacokinetic analysis
A non-compartmental model was used to plot the plasma
The in vivo pharmacokinetic study was performed in male concentration–time values using WinNonlin software (professional
Sprague-Dawley rats (220 ± 10 g). The experimental protocols and edition, version 2.1; Pharsight Corporation, Mountain View, CA,
animal care were in accordance with the protocols laid by Insti- USA). Pharmacokinetic parameters included the elimination rate
tutional Animal Ethical Committee, Yeungnam University, South (Kel ), half-life (t1/2 ), maximum plasma concentration (Cmax ), time
T. Ramasamy et al. / Colloids and Surfaces B: Biointerfaces 146 (2016) 152–160 155

of Cmax (Tmax ), mean retention time (MRT) and area under the
plasma concentration–time curve (AUC).

2.14. In vivo antitumor efficacy study

The experimental protocols and animal care were in accordance


with the protocols laid by Institutional Animal Ethical Committee,
Yeungnam University, South Korea. Antitumor efficacy of formu-
lations was studied on 7-week old female BALB/c nude mice.
The mice were maintained on 12-h daylight cycle with access
to food and water. The A-549 cell suspension (5 × 106 cells in
0.1 mL PBS) was subcutaneously injected into the right flanks of
the mice. Prior to the study, the mice were equally divided into five
groups with 6 mice in each group. Mice in four groups received
free MTX, free DOX, MTX-PCM, and DOX-PCM respectively, at a
fixed dose of 5 mg/kg. Each mouse received 100 ␮L of the formula-
tion administered. The untreated group was observed as control.
After the appropriate tumor volume was attained, formulations
were injected via tail vein (3 times with a gap of 3 days between
each injection). The tumor volume was measured (V = 0.5 × longest
diameter × shortest diameter) using a Vernier caliper. The body
weight of mice was monitored in order to observe the safety pro-
file of the formulations. At the end of the experiment, mice were
sacrificed according to the institutional ethical guidelines. Tumors
were surgically removed and weighed individually.

2.15. Statistical analysis

Student’s t-test was used to evaluate the statistical signifi-


cance of differences between formulations. Values were reported
as mean ± standard deviation (SD) and the data were considered
statistically significant at p < 0.05.

3. Results and discussion

3.1. Preparation of drug-loaded PCM

The present study aimed at investigating the interactions of dif-


ferent cationic drugs with the anionic block polymer PEO-b-PAA.
The primary amino group of DOX (-NH2 ) and two secondary amino
groups (-NH-) of MTX are responsible for electrostatic interactions
with the ionized carboxyl group (pKa ∼ 5) of PEO-b-PAA [11]. There-
fore, pH-responsiveness of individual drugs and their ability to form
drug-loaded PCM were studied in detail. The PCM were formed at
two different charge ratios (R = 0.25 and 0.5) of MTX and DOX to
carboxylate groups (R = [drug]/[COO− ]). The schematic illustration
Fig. 2. Effect of pH on (A) hydrodynamic particle size and (B) ␨-potential at different
of formation of PCM is presented in Fig. 1.
feeding ratios (R = 0.25 and 0.5).
As shown in Fig. 2, the PCM were prepared with both drugs at
various pH conditions. The PCM were formed by the immobilization
of weakly basic drugs (MTX and DOX) into the cores of PEO-b-PAA,
a weak polyacid, via a strong electrostatic interaction. As expected,
we observed pH-sensitive behavior of PCM. Compared to PCM par-
ticle size at pH 6, particle size reduced remarkably with the increase charge of the PAA block results in stable PCM with highly hydropho-
in pH (DOX-PCM). Consistently, ␨-potential of PCM decreased as bic core and high payload capacity [13]. Appreciable hydrophobic
pH increased, indicating that higher pH favors the ionization of the core and PEG shell on the surface stabilize the PCM in systemic
polymer block resulting in complexation of drugs. The ␨-potential conditions. Specifically, MTX-PCM exhibited a smaller particle size
decreased in the entire pH range studied. A possible interpretation compared to that of DOX-PCM. This difference in particle sizes of
is that at lower pH values, owing to partial or insufficient ioniza- cationic drug-based PCM can be attributed to the binding strength
tion of the PAA block, the drug-polymer physical interaction forms of individual drugs to the polymer block. Overall, the size of MTX-
a loose aggregate resulting in larger particle size. Upon increase in PCM and DOX-PCM was less than 100 nm, making these PCM ideal
the pH of the medium, PAA attains maximum ionization resulting for tumor drug delivery. It has been reported that particle smaller
in efficient complexation of drugs. It is worth noting that particle than 200 nm can preferentially accumulate in tumor tissues via
size markedly decreased when the charge ratio was increased from diffusion-mediated passive transport (EPR effect), whereas parti-
R = 0.25 to R = 0.5, indicating the neutralization of the PAA segments cles smaller than 100 nm can penetrate deep in the leaky tumor
due to the electrostatic interaction of MTX and DOX in the PCM. As vasculature (typical pore size 50–100 nm) and are not limited to
expected at higher charge ratios, greater neutralization of negative vascular surface only [14,15].
156 T. Ramasamy et al. / Colloids and Surfaces B: Biointerfaces 146 (2016) 152–160

Fig. 3. TEM images of (A) MTX-PCM and (B) DOX-PCM.

3.2. Morphological and physical state analysis

Regardless of the nature of binding of individual drugs, both


MTX-PCM and DOX-PCM exhibited distinct spherical shaped par-
ticles uniformly dispersed on the TEM grid (Fig. 3A, B). Core-shell
architecture was not observed due to the longitudinal assembly
of polymer chains, but an electron-dense dark core representing
the drug-polymer complex was seen. The particle size observed
in the TEM experiment was smaller than size observed using DLS.
This discrepancy in observed sizes can be attributed to the fact that
DLS measures the hydrodynamic micelle size while TEM captures
the dried state. The physical state of free drugs and drug-loaded
micelles was studied using X-ray diffraction patterns. As shown in
Fig. S1, free DOX showed numerous sharp and intense peaks at sev-
◦ ◦ ◦ ◦ ◦ ◦
eral 2 ␪ scattered angles (12.5 , 16.2 , 17.3 , 21.2 , 22.5 , 25.1 , and

26.2 ) and free MTX showed peaks between 22.5 − 25.5◦ reflecting
its high crystallinity. All characteristic peaks were absent in DOX-
PCM as well as MTX-PCM, indicating the complete incorporation of
drugs. These results suggest the presence of drugs in the amorphous
or molecularly dispersed state [16].

3.3. Drug loading and in vitro release study

Both the DOX-PCM and MTX-PCM exhibited a high entrapment


efficiency of more than 90% with an active drug loading of ∼ 45%
w/w for MTX (MTX-PCM) and ∼ 70% for DOX (DOX-PCM) at R = 0.5.
The release study of MTX and DOX from MTX-PCM and DOX-PCM
was performed in phosphate-buffered saline (pH 7.4) and acetate-
buffered saline (pH 5.0) to simulate the physiological and tumor
pH conditions. As evident from Fig. 4, MTX-PCM and DOX-PCM
exhibited different release profiles at both pH conditions. MTX-
PCM exhibited a sustained release profile throughout the study
period with no initial burst. DOX-PCM, on the other hand, exhibited
a biphasic release pattern. DOX-PCM exhibited a faster release pro-
file during the initial time interval (10–12 h), but showed a slower
release later on (48 h). For instance, in the case of R = 0.5, ∼ 9% of
the drug was released from MTX-PCM after 12 h and approximately
25% of the drug was released by the end of 48 h at pH 7.4. In con-
trast, ∼ 30% of the drug was released from DOX-PCM during the Fig. 4. Release profiles of (A) MTX and (B) DOX from MTX-PCM and DOX-PCM at
first 12 h, while the total release was ∼ 38% at the end of the study pH 5.0 and pH 7.4. MTX-PCM and DOX-PCM were prepared at pH 7.0. The study was
carried out in phosphate-buffered saline (pH 7.4) and acetate-buffered saline (pH
period. A similar trend was observed in release media at pH 5.0
5.0) at 37 ◦ C.
wherein MTX was released in a continuous fashion (monopha-
sic), while DOX was released in a biphasic manner. The difference
in release patterns could be attributed to the charge density and
binding affinity of individual drugs towards the anionic PAA block.
T. Ramasamy et al. / Colloids and Surfaces B: Biointerfaces 146 (2016) 152–160 157

Fig. 5. (A) In vitro cellular uptake of (a) MTX and MTX-PCM and (b) DOX and DOX-PCM in SCC-7 cancer cells. The cellular uptake experiment was performed by incubating
the formulations at different time points. (B) Annexin V-FITC/PI based apoptosis assay in SCC-7 cancer cells. The drug treated cells were stained with Annexin V-FITC and PI
and evaluated using flow cytometry.

MTX, with two secondary amino groups could be expected to have molecules trapped in the core of PCM prepared with R = 0.5 has a
stronger binding affinity for the polymer than DOX with a single greater chance to release quickly in the media than the fewer drug
primary amino group. molecules from the PCM prepared with R = 0.25. Furthermore, at
Another significant observation was that MTX and DOX were the low feeding ratio (R = 0.25), considerable negative charges are
released faster in acidic pH (pH 5.0) than in physiological pH (pH still available on the PAA chain that will further induce strong elec-
7.4). For example, approximately 55% of MTX was released from trostatic interactions between drugs and the polymer leading to
MTX-PCM at acidic pH, while only ∼ 25% of the drug was released slower release rates.
at physiological pH after 48 h at a feeding ratio of R = 0.5. A similar
trend was observed in the case of DOX-PCM, wherein ∼ 62% of the
drug was released in acidic media comparing to ∼ 38% DOX release 3.4. Cellular uptake patterns of DOX-PCM and MTX-PCM
after 48 h in basic media. The accelerated release of drugs at acidic
pH can be attributed to the protonation of carboxylic groups of the The cellular uptake behavior of free drugs and drug-loaded PCM
PAA block in the micelles [11]. was investigated in SCC-7 cancer cells using FACS [18,19]. As shown
In general, it is interesting to note that the release rate was in Fig. 5A, free DOX and MTX showed a higher cellular uptake com-
higher from PCM prepared at a feeding ratio of R = 0.5 than from pared to drug-loaded PCM. The higher cellular uptake of free drugs
those prepared with R = 0.25. For example, ∼ 14% of MTX was was attributed to the simple diffusion of drugs to the intracellular
released from MTX-PCM with R = 0.25 and ∼ 21% was released from environment, whereas micellar nanocarriers could only be inter-
MTX-PCM with R = 0.5 during the first 8 h at pH 5.0. A similar trend nalized by the cells through endocytosis. The mean fluorescent
was observed at pH 7.4: ∼ 17% of DOX was released from DOX-PCM intensity (MFI) of free DOX was greater compared to DOX-PCM and
with R = 0.25 and ∼ 23% was released from PCM with R = 0.5 dur- similarly, MFI of MTX was greater compared to that of MTX-PCM
ing the first 8 h. This difference in release can be attributed to the after 60-min incubation in SCC-7 cancer cells. Consistently, DOX-
binding and localization of the drugs in the core of PCM. A high PCM and MTX-PCM showed a typical time-dependent behavior due
loading capacity of PCM at R = 0.5 accounts for the larger presence to the presence of an endocytosis process within the system (Fig.
of drugs at the core-shell interface from where the drugs can rapidly S2). We have observed that the nanocarriers primarily accumu-
diffuse into the release medium [17]. The greater number of drug late in the cytoplasmic region where the drug is liberated after the
158 T. Ramasamy et al. / Colloids and Surfaces B: Biointerfaces 146 (2016) 152–160

degradation of delivery systems. These results indicate an efficient


internalization of PCM in this cell line.

3.5. Apoptosis assay

The externalization of phosphatidylserine during apoptosis of


cancer cells was evaluated by annexin-V/PI staining (Fig. 5B). The
results showed that DOX induced a significant increase in Annexin
V-positive and Annexin V + PI-positive cells, corresponding to
early and late apoptotic cells, respectively. The MTX, however,
induced apoptosis as well as necrosis of cancer cells. Importantly,
drug-loaded PCM induced higher apoptosis rates of cancer cells
compared to free drugs. For example, MTX-PCM induced ∼ 25%
of cell apoptosis compared to ∼ 15% induced by free MTX. Simi-
larly, DOX-PCM induced ∼ 34% of cell apoptosis compared to ∼ 25%
induced by free DOX. Higher cellular apoptosis rates induced by
PCM could be attributed to the sustained release of therapeutic
cargo in the intracellular environment. It should be noted that DOX-
based therapy was more effective in inducing anticancer activity
than MTX-based therapy.

3.6. Pharmacokinetic analysis

The plasma concentration-time profiles of free drugs, MTX-PCM,


and DOX-PCM following single dose administration are presented
in Fig. 6. As shown, free MTX and free DOX were cleared from
the systemic compartment within 4–6 h of intravenous admin-
istration. Linear pharmacokinetic profiles of MTX and DOX were
consistent with previous reports. As expected, PCM formulations
significantly enhanced the blood circulation of both MTX and DOX.
Both anticancer drugs maintained significantly higher plasma con-
centrations for 24 h. Importantly, MTX-PCM showed prolonged
blood circulation, compared to DOX-PCM. After 12 h, the plasma
concentration of MTX from PCM was 1.824 ± 0.801 ␮g/ml com-
pared to plasma concentration of only 0.576 ± 0.389 ␮g/mL for
DOX. The final concentrations of MTX and DOX released from PCM
were 1.258 ± 0.392 ␮g/mL and 0.176 ± 0.151 ␮g/mL, respectively.
The respective pharmacokinetic parameters of different formu-
lations are presented in Table 1. Consistent with previous reports,
free drugs exhibited short t1/2 , high Kel , and low AUCall . Although
both PCM formulations improved the systemic performance of
drugs, they markedly differ among themselves. For instance, MTX-
PCM improved the AUCall of MTX 4-fold compared to a 3-fold
increase by DOX-PCM for DOX. Similarly, MTX-PCM had an approxi- Fig. 6. Plasma concentration-time profiles of MTX and DOX after intravenous
administration of free drugs or drug-loaded PCM to rats at a dose of 5 mg/kg. Each
mately 5-fold (14.79 ± 4.89 h) higher t1/2 than MTX, compared with
value represents the mean ± SD (n = 4). Drug-loaded PCM were prepared at R = 0.5
2.5-fold higher t1/2 of DOX-PCM (4.82 ± 0.83 h) in relation to DOX. and pH 7.0.
Notably, Kel of the free drugs was reduced 5-fold by MTX-PCM and
2-fold by DOX-PCM. With regard to all pharmacokinetic parame-
ters, MTX-PCM showed 2-fold higher performance than DOX-PCM. 3.7. In vivo antitumor efficacy
These findings indicate the remarkable blood circulation potential
of MTX-PCM compared to that of DOX-PCM. The difference in the The prolonged blood circulation and controlled release pro-
circulatory performance of the two PCM formulations is attributed files of drug-loaded PCM were expected to contribute to their
to their physiological stability [20]. Previously, we have shown that superior antitumor efficacy. The antitumor efficacy of individual
DOX-PCM have lower salt stability than MTX-PCM. Two secondary formulations was investigated in A-549 cancer cells xenografted
amino groups confer on MTX a stronger binding affinity for the on BALB/c nude mice. Free MTX, free DOX, MTX-PCM, and DOX-
polymer, compared to DOX with its single primary amino group PCM were intravenously injected into the tumor bearing mice at
[11,21]. a fixed dose of 5 mg/kg. As shown in Fig. 7A, tumors rapidly grew
Many inferences can be drawn from this experiment. First, the in the untreated control group, but their growth was significantly
binding affinity of the cationic drug to the polymer determines its suppressed in groups treated with free drugs as well as drug-
blood circulation potential; second, based on the binding strength, loaded PCM. Notably, both MTX-PCM and DOX-PCM significantly
the release of the drug will be sustained or faster; third, the greater suppressed tumor growth. In vitro cytotoxicity assays revealed the
the binding strength, the greater the in vivo performance of drug- IC50 values for individual formulations. The IC50 values of MTX and
loaded nanocarriers in the physiological environment [22–25]. MTX-PCM were found to be 0.85 ␮g/ml and 0.94 ␮g/ml, respec-
T. Ramasamy et al. / Colloids and Surfaces B: Biointerfaces 146 (2016) 152–160 159

Table 1
Pharmacokinetic parameters of MTX and DOX after IV administration of free drug or drug-loaded PCM to rats.

Parameters MTX MTX-PCM DOX DOX-PCM

Kel (h−1 ) 0.25 ± 0.049 0.052 ± 0.016 0.36 ± 0.054 0.15 ± 0.022
t1/2 (h) 2.88 ± 0.514 14.79 ± 4.895 1.93 ± 0.325 4.82 ± 0.823
AUCall (h ␮g ml−1 ) 13.18 ± 1.497 47.85 ± 14.82 8.18 ± 1.27 27.66 ± 8.85
AUCinf (h ␮g ml−1 ) 14.42 ± 2.325 78.32 ± 35.22 8.72 ± 1.72 29.03 ± 9.78
Cl (␮g ml−1 h−1 ) 141.87 ± 20.59 31.94 ± 15.12 237.44 ± 41.32 79.82 ± 32.92
AUMC (␮g ml−1 h) 42.78 ± 6.34 309.71 ± 257.49 18.66 ± 3.78 162.82 ± 67.68
MRT (h) 3.23 ± 0.135 9.58 ± 0.615 2.26 ± 0.118 5.71 ± 0.72

results show that despite the difference in circulatory performance


and drug release patterns between MTX-PCM and DOX-PCM their
therapeutic effect is similar. It should be noted that DOX-PCM were
more effective than free DOX in suppressing tumors, whereas anti-
tumor efficacy of MTX-PCM and free MTX was observed to be
similar. This was attributed to the high toxicity of MTX on mice,
which resulted in overall body weakness that affected the tumor
tissue as well. The enhanced tumor regression caused by PCM for-
mulations can be attributed to the prolonged half-life of anticancer
drugs, reduced elimination of individual drugs, and most impor-
tantly to the preferential accumulation of nanocarriers in the tumor
tissue due to the EPR effect [26–28].
The toxicity of formulations was evaluated using mice body
weight (Fig. 7B). As shown, free MTX caused an approximately 30%
decrease in body weight indicating its severe drug-related toxicity.
MTX-PCM, however, greatly reduced MTX toxicity in systemic cir-
culation. This could be due to the fact that encapsulation of MTX in
the PCM reduced the random exposure of normal tissues to it and
increased MTX’s passive accumulation in tumor tissues, thereby
reducing the undesirable side effects [29,30]. DOX-PCM did not
exhibit any body weight reduction.

4. Conclusion

In summary, cationic drugs-loaded PCM were prepared and


evaluated in terms of physicochemical and in vivo parameters. Both
MTX-PCM and DOX-PCM displayed spherical nanosized particles
with uniform dispersity indices. MTX-PCM and DOX-PCM exhibited
different release profiles under all pH conditions studied. MTX-PCM
exhibited a monophasic release pattern with no initial burst, while
DOX-PCM exhibited a biphasic release pattern. Interestingly, drug
release rates were higher from PCM prepared at a feeding ratio of
R = 0.5 than from those prepared with R = 0.25. DOX-PCM showed
a higher cellular uptake compared to MTX-PCM in SCC-7 cancer
cells; consistently DOX-PCM induced higher apoptosis rates of can-
cer cells than MTX-PCM. In contrast, MTX-PCM showed prolonged
blood circulation compared to DOX-PCM. MTX-PCM improved the
AUCall of MTX 4-fold compared to a 3-fold increase by DOX-PCM for
DOX. Similarly, MTX-PCM had a 5-fold higher t1/2 than MTX, while
DOX-PCM increased the DOX t1/2 2.5-fold. However, both MTX-
PCM and DOX-PCM suppressed tumor growth to the same levels as
their respective free drugs. Taken together, our results show that
nature of interactions of cationic drugs with the polyionic copoly-
mer can have a tremendous influence on the biological performance
Fig. 7. Effect of drug-loaded PCM on (A) tumor growth and (B) body weight in A-549 of delivery systems.
xenograft-bearing female BALB/c nude mice (n = 6 per group). Each formulation was
administered three times at three day intervals. Drug-loaded PCM were prepared at
R = 0.5 and pH 7.0. Conflict of interests

The authors declare no conflict of interest in this work.


tively, while the IC50 values of DOX and DOX-PCM were 1.68 ␮g/ml
and 2.13 ␮g/ml, respectively. While a definite difference in blood
circulation was observed between MTX-PCM and DOX-PCM in the Acknowledgements
pharmacokinetic study, no significant difference in antitumor effi-
cacy could be detected. Both MTX-PCM and DOX-PCM inhibited This work was supported by the 2015 Yeungnam University
tumor growth to the same level throughout the study period. These Research Grant.
160 T. Ramasamy et al. / Colloids and Surfaces B: Biointerfaces 146 (2016) 152–160

Appendix A. Supplementary data [16] B. Gupta, B.K. Poudel, S. Pathak, J.W. Tak, H.H. Lee, J.H. Jeong, H.G. Choi, C.S.
Yong, J.O. Kim, Effects of formulation variables on the particle size and drug
encapsulation of imatinib-loaded solid lipid nanoparticles, AAPS
Supplementary data associated with this article can be found, in PharmSciTech (2015).
the online version, at http://dx.doi.org/10.1016/j.colsurfb.2016.06. [17] T. Ramasamy, J.H. Kim, J.Y. Choi, T.H. Tran, H.G. Choi, C.S. Yong, J.O. Kim, pH
004. sensitive polyelectrolyte complex micelles for highly effective combination
chemotherapy, J. Mater. Chem. B 2 (2014) 6324.
[18] X. Qi, Y. Chen, Z. Sun, X. Ma, W. Guo, Y. Lu, Duan, Cytotoxicity and cellular
References uptake evaluation of mitoxantrone-loaded poly(lactic acid-co-lysine)
arginine–glycine–aspartic acid nanoparticles, J. Appl. Polym. Sci. 119 (2011)
[1] M.M. Gottesman, T. Fojo, S.E. Bates, Multidrug resistance in cancer: role of 1011–1015.
ATP dependent transporters, Nat. Rev. Cancer 2 (2002) 48. [19] R. Zeng, A.M. Morgenstern, Nyström, Nanoparticle-directed sub-cellular
[2] A.T. Fojo, K. Ueda, D.J. Slamon, D.G. Poplack, M.M. Gottesman, I. Pastan, localization of doxorubicin and the sensitization breast cancer cells by
Expression of a multidrug-resistance gene in human-tumors and tissues, Proc. circumventing GST-mediated drug resistance, Biomaterials 35 (2014)
Natl. Acad. Sci. U. S. A. 84 (1987) 265. 1227–1239.
[3] G. Riess, Micellization of block copolymers, Prog. Polym. Sci. 28 (2003) [20] H.B. Ruttala, Y.T. Ko, Liposome encapsulated albumin-paclitaxel nanoparticle
1107–1170. for enhanced antitumor efficacy, Pharm. Res. 32 (2015) 1002–1016.
[4] J.F. Cohy, Block copolymer micelles, Adv. Polym. Sci. 190 (2005) 65–136. [21] J.O. Kim, H.S. Oberoi, S. Desale, A.V. Kabanov, T.K. Bronich, Polypeptide
[5] H. Maeda, The enhanced permeability and retention (EPR) effect in tumor nanogels with hydrophobic moieties in the cross-linked ionic cores:
vasculature: the key role of tumor-selective macromolecular drug targeting, synthesis, characterization and implications for anticancer drug delivery, J.
Adv. Enzyme Regul. 41 (2001) 189–207. Drug Target. 21 (2013) 981–993.
[6] J.O. Kim, A.V. Kabanov, T.K. Bronich, Polymer micelles with cross-linked [22] T.H. Tran, T. Ramasamy, D.H. Truong, B.S. Shin, H.G. Choi, C.S. Yong, J.O. Kim,
polyanion core for delivery of a cationic drug doxorubicin, J. Control. Rel. 138 Development of vorinostat-loaded solid lipid nanoparticles to enhance
(2009) 197–204. pharmacokinetics and efficacy against multidrug-resistant cancer cells,
[7] J.O. Kim, T. Ramasamy, C.S. Yong, N.V. Nukolova, T.K. Bronich, A.V. Kabanov, Pharm. Res. 31 (2014) 1978–1988.
Cross-linked polymeric micelles based on block ionomer complexes, [23] T.H. Tran, J.Y. Choi, T. Ramasamy, D.H. Truong, C.N. Nguyen, H.G. Choi, C.S.
Mendeleev Commun. 23 (2013) 179–186. Yong, J.O. Kim, Hyaluronic acid-coated solid lipid nanoparticles for targeted
[8] A.V. Kabanov, T.K. Bronich, V.A. Kabanov, K. Yu, A. Eisenberg, Soluble delivery of vorinostat to CD44 overexpressing cancer cells, Carbohydr. Polym.
stoichiometric complexes from poly(N-ethyl-4-vinylpyridinium) cations and 114 (2014) 407–415.
poly(ethylene oxide)-block-polymethacrylate anions, Macromolecules 29 [24] T. Ramasamy, Z.S. Haidar, Formulation, characterization and cytocompatibility
(1996) 6797–6802. evaluation of novel core–shell solid lipid nanoparticles for the controlled and
[9] T.K. Bronich, A.M. Popov, A. Eisenberg, V.A. Kabanov, A.V. Kabanov, Effects of tunable delivery of a model protein, J. Bionanosci. 5 (2011) 143–154.
block length and structure of surfactant on self-assembly and solution [25] T. Ramasamy, T.H. Tran, J.Y. Choi, H.J. Cho, J.H. Kim, J.H. Choi, C.S. Yong, J.O.
behavior of block ionomer complexes, Langmuir 16 (2000) 481–489. Kim, Layer-by-layer coated lipid?polymer hybrid nanoparticles designed for
[10] T. Ramasamy, T.H. Tran, H.J. Cho, J.H. Kim, Y.I. Kim, J.Y. Jeon, H.G. Choi, C.S. use in anticancer drug delivery, Carb. Polym. 102 (2014) 653–661.
Yong, J.O. Kim, Chitosan-based polyelectrolyte complexes as potential [26] J.U. Assumpçao, M.L. Campos, M.A. Ferraz Nogueira Filho, K.C. Pestana, H.M.
nanoparticulate carriers: physicochemical and biological characterization, Baldan, T.P. Formariz Pilon, et al., Biocompatible microemulsion modifies the
Pharm. Res. 31 (2014) 1302–1314. pharmacokinetic profile and cardiotoxicity of doxorubicin, J. Pharm. Sci. 102
[11] T. Ramasamy, J.H. Kim, H.G. Choi, C.S. Yong, J.O. Kim, Novel dual drug-loaded (2013) 289.
block ionomer complex micelles for enhancing the efficacy of chemotherapy [27] D.S. Alberts, Y.M. Peng, G.T. Bowden, W.S. Dalton, C. Mackel, Pharmacology of
treatments, J. Biomed. Nanotech. 10 (2014) 1304–1312. mitoxantrone: mode of action and pharmacokinetics, Invest. New Drugs 3
[12] B. Manocha, A. Margaritis, Controlled release of doxorubicin from (1985) 101–107.
doxorubicin/-polyglutamic acid ionic complex, J. Nanomat. 2010 (2010) [28] J.H. Kim, Y.S. Kim, K. Park, S. Lee, H.Y. Nam, K.H. Min, et al., Antitumor efficacy
(Article ID 780171). of cisplatin-loaded glycol chitosan nanoparticles in tumor-bearing mice, J.
[13] T. Ramasamy, Z.S. Haidar, T.H. Tran, J.Y. Choi, J.H. Jeong, B.S. Shin, H.G. Choi, Control. Rel. 127 (2008) 41–49.
C.S. Yong, J.O. Kim, Layer-by-layer assembly of liposomal nanoparticles with [29] T. Ramasamy, H.B. Ruttala, J.Y. Choi, T.H. Tran, J.H. Kim, S.K. Ku, H.G. Choi, C.S.
PEGylated polyelectrolytes enhances systemic delivery of multiple anticancer Yong, J.O. Kim, Engineering of a lipid-polymer nanoarchitectural platform for
drugs, Acta Biomat. 10 (2014) 5116–5127. highly effective combination therapy of doxorubicin and irinotecan, Chem.
[14] D.W. Kim, T. Ramasamy, J.Y. Choi, J.H. Kim, C.S. Yong, J.O. Kim, H.G. Choi, The Commun. 51 (2015) 5758.
influence of bile salt on the chemotherapeutic response of docetaxel loaded [30] T. Ramasamy, J.Y. Choi, H.J. Cho, S.K. Umadevi, B.S. Shin, H.G. Choi, C.S. Yong,
thermosensitive nanomicelles, Int. J. Nanomed. 9 (2014) 3815–3824. J.O. Kim, Polypeptide-based micelles for delivery of irinotecan:
[15] B. Gupta, B.K. Poudel, T.H. Tran, R. Pradhan, H.J. Cho, J.H. Jeong, B.S. Shin, H.G. physicochemical and in vivo characterization, Pharm. Res. 32 (2015)
Choi, C.S. Yong, J.O. Kim, Modulation of pharmacokinetic and cytotoxicity 1947–1956.
profile of imatinib base by employing optimized nanostructured lipid carriers,
Pharm. Res. 32 (2015) 2912–2927.

You might also like