Subscriber access provided by the Henry Madden Library | California State University, Fresno
Article
N-deoxycholic acid-N,O-hydroxyethyl chitosan with a sulfhydryl modification to enhance the oral absorptive efficiency of paclitaxel Yanfang Yu, Meirong Huo, Ying Fu, Wei Xu, Han Cai, Lingling Yao, Qinyu Chen, Yan Mu, Jianping Zhou, and Tingjie Yin Mol. Pharmaceutics, Just Accepted Manuscript • DOI: 10.1021/acs.molpharmaceut.7b00662 • Publication Date (Web): 23 Oct 2017 Downloaded from http://pubs.acs.org on October 24, 2017
Just Accepted “Just Accepted” manuscripts have been peer-reviewed and accepted for publication. They are posted online prior to technical editing, formatting for publication and author proofing. The American Chemical Society provides “Just Accepted” as a free service to the research community to expedite the dissemination of scientific material as soon as possible after acceptance. “Just Accepted” manuscripts appear in full in PDF format accompanied by an HTML abstract. “Just Accepted” manuscripts have been fully peer reviewed, but should not be considered the official version of record. They are accessible to all readers and citable by the Digital Object Identifier (DOI®). “Just Accepted” is an optional service offered to authors. Therefore, the “Just Accepted” Web site may not include all articles that will be published in the journal. After a manuscript is technically edited and formatted, it will be removed from the “Just Accepted” Web site and published as an ASAP article. Note that technical editing may introduce minor changes to the manuscript text and/or graphics which could affect content, and all legal disclaimers and ethical guidelines that apply to the journal pertain. ACS cannot be held responsible for errors or consequences arising from the use of information contained in these “Just Accepted” manuscripts.
Molecular Pharmaceutics is published by the American Chemical Society. 1155 Sixteenth Street N.W., Washington, DC 20036 Published by American Chemical Society. Copyright © American Chemical Society. However, no copyright claim is made to original U.S. Government works, or works produced by employees of any Commonwealth realm Crown government in the course of their duties.
Page 1 of 32
1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60
Molecular Pharmaceutics
1
N-deoxycholic acid-N,O-hydroxyethyl chitosan with a sulfhydryl modification to enhance the oral absorptive
2
efficiency of paclitaxel
3
Yanfang Yu,1 Meirong Huo,1 Ying Fu, Wei Xu, Han Cai, Lingling Yao, Qingyu Chen,Yan Mu, Jianping Zhou*, Tingjie Yin*
4 5
a
6
Tongjiaxiang, Nanjing 210009, China
7
b
State Key Laboratory of Natural Medicines, Department of Pharmaceutics, China Pharmaceutical University, 24
Department of Pharmacy, Shandong Provincial QianFoshan Hospital, Shandong University, Jinan 250014, China.
8 9
ABSTRACT
10
Currently, the most prominent barrier to the success of orally delivered paclitaxel (PTX) is the extremely limited
11
bioavailability of delivered therapeutic. In light of this issue, an amphiphilic sulfhydrylated N-deoxycholic
12
acid-N,O-hydroxyethyl chitosan (TGA-DHC) was synthesized to improve the oral bioavailability of PTX. Firstly,
13
TGA-DHC demonstrated substantial loading of PTX into the inner hydrophobic core. A desirable enhancement in the
14
bioavailability of PTX by TGA-DHC was verified by pharmacokinetic studies on rats against Taxol® and
15
non-sulfhydrylated DHC micelles. Moreover, cellular uptake studies revealed significant accumulation of TGA-DHC
16
micelles encapsulating PTX or rhodamine-123 into Caco-2 cells via clathrin/caveolae-mediated endocytosis and inhibition
17
of P-gp efflux of substrates. The results of the Caco-2 transport study further confirmed the mechanistic basis of TGA-DHC
18
efficacy; which was attributed to permeabilized tight junctions, clathrin-mediated transcytosis across the endothelium and
19
inhibition of P-gp. Finally, in vitro mucoadhesion investigations on freshly excised rat intestine intuitively confirmed
20
increased intestinal retention of drug-loaded TGA-DHC through thiol-mediated mucoadhesion. TGA-DHC has
21
demonstrated the capability to overcome what is perhaps the most prominent barrier to oral PTX efficacy; low
22
bioavailability, and serves as a prominent platform for oral delivery of P-gp substrates.
23 24
KEYWORDS: paclitaxel, sulfhydrylated N-deoxycholic acid-N,O-hydroxyethyl chitosan, micelles, oral delivery,
25
mucoadhesion
26 27
1. INTRODUCTION
28
Paclitaxel (PTX) is an extensively used, clinical front-line therapeutic against a wide spectrum of solid cancers.
29
Mechanistically, PTX effects over-stabilization of cellular microtubules, preventing mitosis. Therefore, cell apoptosis occurs
30
at the late G2/M phase.1 In its commercially available formulation, Taxol® (Bristol-Myers Squibb, New York, NY, USA),
31
PTX is formulated in a 50:50 (v/v) mixture of Cremophor EL and dehydrated ethanol, further diluted in 0.9% saline solution 1
ACS Paragon Plus Environment
Molecular Pharmaceutics
1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60
Page 2 of 32
32
forintravenous (i.v.) administration. However, any therapeutic delivered by i.v. administration comes with an increased risk
33
of catheter-related infection, potential thrombosis, and extravasation. Furthermore, in the case of Taxol®, Cremophor EL has
34
been known to be the cause of hypersensitivity, neurotoxicity, nephrotoxicity, and cardiotoxicity during the course of
35
treatment.2 Consequently, the delivery of PTX through safer, alternate methods has remained an area of active investigation.
36
Oral delivery of PTX has appealed over i.v. routes due to factors such as patient convenience, compliance, and the
37
relative safety of the oral route in terms of minimizing infection. Yet, orally delivered Taxol® therapy is sparingly used due
38
to its low oral bioavailability (< 10%).3 Biologically, the low availability is mainly attributed to the poor solubility and
39
dissolution of PTX in gastrointestinal fluids. Subsequent hepatic metabolism of drug by cytochrome P450 metabolic
40
enzymes (CYP450), rapid clearance by mucus, low permeability across the epithelium, and efflux of PTX by multidrug
41
transporter P-glycoprotein (P-gp) all contribute to the insufficient bioavailability of orally delivered Taxol®.4,5
42
The advent of nanotechnology has revealed a wide range of prospects for efficient oral drug delivery. Recently, numerous
43
nanocarriers including liposomes, microemulsions, cyclodextrins, nanogels and polymeric micelles have been developed to
44
improve oral PTX bioavailability.6,7 In particular, polymeric micelles (PMs) possess appealing properties such as nanoscopic
45
dimensions and distinctive core-shell structures.8,9 Insoluble molecules such as PTX can be easily solubilized through an
46
effective encapsulation into the hydrophobic inner core of PMs. In addition, the hydrophilic outer shell of PMs confers
47
aqueous solubility and steric stability; protecting PTX from inactivation in gastrointestinal fluids or CYP450 recognition.
48
Resultantly, PMs have attracted great attention as effective oral PTX delivery carriers. Yet, a PM designed for oral PTX
49
delivery must still overcome PTX effluxby P-gp after release from the delivery carrier, rapid mucosal clearance and low
50
epithelium permeability.10
51
Chitosan (CS), a cationic polysaccharide derived from alkaline N-deacetylation of chitin, has garnered interest as an oral
52
delivery carrier owning to its well-known biocompatibility, biodegradability and adhesion properties to mucosal
53
membranes.11 CS and its derivatives were also reported to enhance oral drug absorption through opening of tight junctions
54
(TJ) to mediate paracellular permeation and further promote P-gp inhibition through P-gp-independent internalization in the
55
form of integral drug-loaded micelles and P-gp ATPase inhibition by free polymers.12 Therefore, chitosan based PMs offer a
56
multitude of advantages for transport of PTX across the epithelium while overcoming the issue of P-gp based efflux.
57
To further enhance the efficacy of orally administered PMs, the influence of particle clearance by the mucus lining must
58
also be taken into consideration. The slippery lining of many epithelial tissues is quickly renewed, and functions to rapidly
59
remove foreign matter.13 In the case of drug delivery, however, mucosal renewal presents a prominent pathway for reduced
60
bioavailability. In light of this issue, thiolated polymers, often referred to as thiomers, have emerged as a promising mucosal
61
adhesion-enhancing excipient for oral drug delivery.14 Such polymers are capable of adhering to the mucosa through
62
formation of covalent disulfide linkage between their free sulfhydryl group and the cysteine-rich mucosal glycoprotein; 2
ACS Paragon Plus Environment
Page 3 of 32
1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60
Molecular Pharmaceutics
63
leading to an enhanced mucus retention. Furthermore, thiomers demonstrate athiol mediated P-gp and CYP450 inhibitory
64
activity.15,16 Investigations have confirmed that the absorption of the P-gp-substrate, Rhodamine 1223 (Rho-123), across the
65
intestine was increased by 3-fold in the presence of CS-4-thiobutylamidine (CS-TBA) over an untreated
66
control.17Additionally, poly (acrylic acid)-cysteine conjugated with reduced glutathione (GSH) prepared by Iqbal et al.
67
exhibited a 7.4-fold increase in PTX oral absorption compared with the buffer solution control.18
68
Considering the advancements offered by thiolated, chitosan based PMs for enhancing the bioavailability of orally
69
delivered therapeutics, we herein proposed an amphiphilic sulfhydrylated N-deoxycholic acid-N,O-hydroxyethyl chitosan
70
(TGA-DHC)-based polymeric micelle to improve the oral bioavailability of PTX. Through an effective solubilization by
71
TGA-DHC, PTX was entrapped in the hydrophobic cores of TGA-DHC micelles and protected from CYP450 degradation
72
in the gastrointestinal tract. Subsequently, PTX-loaded TGA-DHC micelles demonstrated thiol-mediated mucosal adhesion
73
for increased intestinal retention, followed by a P-gp-independent internalization in their intact core/shell form by
74
enterocytes or M cells. Furthermore, TGA-DHC polymers permitted opening of tight junctions for paracellular permeation
75
while inhibiting P-gp efflux to enhance absorption of released PTX from TGA-DHC PMs (Scheme 1). The present study
76
investigated the basic physicochemical characteristics of micelles with or without PTX encapsulation (TGA-DHC and
77
PTX-TGA-DHC, respectively), compared the oral bioavailability of PTX-TGA-DHC micelles with non-sulfhydrylated
78
PTX-DHC micelles and Taxol®. Finally, the mechanism of TGA-DHC on oral absorptive enhancement was studied.
79 80
Scheme 1. Scheme of the construction of PTX-TGA-DHC and its effect on improving the oral absorption of PTX including
81
mucous adhesion (A), transcytosis across endothelium (B), paracellular transport through the opening of tight junctions (C),
82
and inhibition of PTX efflux by P-gP (D).
83 3
ACS Paragon Plus Environment
Molecular Pharmaceutics
1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60
84
Page 4 of 32
2. MATERIAL AND METHODS
85
2.1. Materials. Chitosan (100 kDa) with deacetylation degree of 90.75% was purchased from the Aoxing Biotechnology
86
Co., Ltd. (Yuhuan, China). N-deoxycholic acid-N,O-hydroxyethyl chitosan (DHC) was synthesized as described in our
87
previous article, and the substitution degree of deoxycholic acid and hydroxyethyl determined by elemental analysis was
88
3.77% and 106.57%, respectively.19 Paclitaxel (PTX) was purchased from Shanghai Sanwei Pharmaceutical Co.Ltd.
89
Verapamil was offered by Hengrui Pharmaceutical Co., Ltd. (Jiangsu, China). Cell Counting Kit-8 (CCK8) was purchased
90
by Dojindo Molecular Technologies, Inc. (Shanghai, China). Rhodamine 123 (R-123) and BCA protein assay kit were
91
purchased from Beyotime Institute of Biotechnology (Shanghai, China). Dulbecco’s modified Eagle medium (DMEM, high
92
glucose), trypsin-EDTA (0.25%), fetal bovine serum, penicillin-streptomycin solution, nonessential amino acid (NEAA)
93
solution, Hank’s buffered salt solution (HBSS) and phosphate buffered saline (PBS), were provided by Thermo Fisher
94
Scientific Inc. (Beijing, China). Chlorpromazine, amiloride and nystatin were purchased from Sigma-Aldrich Co. (Shanghai,
95
China). All other chemicals and reagents were of analytical grade.
96 97
2.2. Animals and Cell Lines. Sprague-Dawley (SD) rats (180-220g) were obtained from Origin Biosciences Inc.
98
(Nanjing, China). All animals were received care incompliance with the National Institute of Health Guide for the Care and
99
Use of Laboratory Animals.
100
Caco-2 cells were obtained from the cell bank of Chinese Academy of Sciences, and frozen at -170 ºC in liquid nitrogen
101
before use. The cells were cultured in complete DMEM with 1% (v/v) NEAA, 100 U/mL penicillin and 100 µg/mL
102
streptomycin at 37 ºC in 5% CO2 atmosphere and 90% relative humidity. The cells were sub-cultivated every 5 days at a
103
split ratio of 1:10.
104 105
2.3. Synthesis of Sulfhydrylated N-deoxycholic acid-N,O-hydroxyethyl Chitosan (TGA-DHC). 250 mg of DHC was
106
dissolved in 25 mL of distilled water with addition of EDC, NHS and thioglycolic acid (TGA) at different molar ratios
107
(nDHC:nTGA:nEDC:nNHS). The reaction was stirred away from light at pH of 3, 4, 5, or 6. After 24 h, the mixture was dialyzed
108
against 5 mM HCl using a dialysis membrane (MWCO: 12-14 kDa) for 72 h, 5 mM HCl containing 1% NaCl for 48 h, 1
109
mM HCl for 28 h, and subsequently lyophilized. The TGA-DHC was obtained and stored at 4 ºC.
110 111
2.4. Characterization of TGA-DHC. Ellman's method was used to estimate the number of thiol groups on TGA-DHC.20
112
0.5 mg of TGA-DHC was dissolved in pH 8.0 PBS (0.5 M) with or without 4% NaBH4 and stirred at 37 ºC for 1 h.
113
Subsequently, 200 µL of 5 M HCl was added to inactivate NaBH4, followed by addition of 100 µL Ellman's reagent. The
114
amount of free and totalthiol groups was quantitated using a standard curve prepared from solutions with a known amount 4
ACS Paragon Plus Environment
Page 5 of 32
1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60
115
Molecular Pharmaceutics
of thiol groups.
116 117
2.5. Preparation of PTX-loaded Micelles. PTX-loaded micelles (PTX-DHC, PTX-TGA-DHC) were prepared by
118
dialysis. PTX (10 mg) was dissolved in 300 µL of dehydrated ethanol while DHC/TGA-DHC was dissolved in 3 mL of
119
distilled water. The PTX solution was added dropwise into the DHC or TGA-DHC solution under constant stirring for 0.5 h.
120
The mixture was dispersed using a probe-type ultrasonicator (JY92-2D; Ningbo Scientz Biotechnology Co., Ltd., China) at
121
100 W for 30 min and dialyzed against distilled water overnight. After filtration by a 0.45 µm filter, the DHC-PTX and
122
TGA-DHC-PTX were obtained and stored at 4 ºC until use.
123
The entrapment efficiency (EE) and drug-loading (DL) were calculated by the following formulas: EE % = DL % =
amount of PTX in micelles × 100 amount of PTX dissolved in ethanol
amount of PTX in micelles × 100 amount of PTX in micelles + amount of conjugates fed initially
124
The amount of PTX was determined by high-performance liquid chromatography (HPLC, Shimadzu LC-2010 system,
125
Kyoto, Japan) with UV detection at 227 nm using a LichrospheTM C18 column (5 µm particle size, 250 mm × 4.6 mm).
126 127
2.6. Characterization of PTX-loaded Micelles. The particle size and zeta potential of PTX-DHC and PTX-TGA-DHC
128
were measured using dynamic light scattering (DLS, Nano-ZS90, Malvern Instruments, UK). The morphology of
129
TGA-DHC-PTX was observed by transmission electron microscopy (TEM, H-600, Hitachi, Japan). PTX-loaded micelles
130
for TEM analysis were negatively stained by phosphotungstic acid (1%, v/v). The efficient encapsulation of PTX into
131
TGA-DHC was demonstrated by wide-angle X-ray diffraction (WAXD) and differential scanning calorimetric (DSC)
132
analysis. WAXD was performed using an XD-3A powder diffraction meter (Bruker, AXS, Germany) with Cu Ka radiation.
133
DSC analysis was performed using NETZSCH DSC 204 equipment with the temperature and heating rate of 40-300 °C and
134
10 °C/min, respectively.
135 136
2.7. In Vitro Drug Release. The PTX release from Taxol®, PTX-DHC and PTX-TGA-DHC micelles was evaluated by
137
dialysis. Taxol®, PTX-DHC and PTX-TGA-DHC micelles containing 1 mg of PTX were dissolved in 1 mL of distilled
138
water and placed into a dialysis bag (MWCO = 12,000-14,000 Da). The whole bag was immersed in 150 mL of artificial
139
gastric juice (pH 1.2) containing 0.1% (w/v) Tween 80 for 2 h, followed by another 24 h of incubation with artificial
140
intestinal fluid (pH 6.8) containing 0.1% (w/v) Tween 80. The experiment was performed in an incubating shaker at 37 ºC
141
and 100 rpm. At predetermined time intervals, 1 mL of release media was removed for HPLC analysis and the whole media
142
was refreshed. All data points represent the average of six readings.
143 5
ACS Paragon Plus Environment
Molecular Pharmaceutics
1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60
Page 6 of 32
144
2.8. Pharmacokinetic Studies. To investigate the enhancement on the oral absorption of PTX by PTX-TGA-DHC, nine
145
rats were divided randomly into the following groups (n = 3): (1) Taxol® (p.o.); (2) PTX-DHC (p.o.); (3) PTX-TGA-DHC
146
(p.o.); (4) Taxol® (i.v.). The intragastric administration dose of PTX was 20 mg/kg, while the i.v. administration dose of
147
PTX was 7 mg/kg. After oral administration of PTX formulations at a total dose of 20 mg/kg, blood samples were collected
148
into heparinized eppendorf tubes at 0.17, 0.33, 0.5, 1, 2, 4, 8, 12, 24, 48 h. Blood samples from animals receiving Taxol®
149
administered intravenously were collected at 0.083, 0.17, 0.33, 0.67, 1, 2, 4, 8, 12 h. The collected samples were centrifuged
150
at 9648 ×g for 10 min, and the plasma was obtained and stored at -20 ºC for HPLC analysis.
151
Plasma concentration of PTX versus time in rats was analyzed using standard non-compartment analysis with the aid of
152
PKSolver, an add-in program for pharmacokinetic and pharmacodynamic data analysis by our group.21 The main
153
pharmacokinetic parameters used in this study were the area under the plasma concentration-time curve (AUC0-∞) for
154
intravenous administration, the area under the concentration-time profilefrom time zero to 48 h (AUC0-48 h) for oral
155
administration, total body clearance (CL), mean residence time (MRT), maximum concentration (Cmax ) and the time to Cmax
156
(Tmax ).
157
Furthermore, the relative bioavailability (F%) of PTX from these oral formulations was calculated as the following
158
equation: F% = (AUC0-48 h × Di.v.)/(AUC0-∞ × Dp.o.) × 100%, where AUC0-48 h was the areaunder the concentration-time
159
profile from time zero to 48 h from the oral administration, while AUC0-∞ was the area under the plasma concentration-time
160
curve from the intravenous administration. Meanwhile, Di.v. and Dp.o. represented the dosage of PTX for the intravenous and
161
oral administration, respectively.
162 163
2.9. In Vitro Cytotoxicity. In vitro cytotoxicity of blank micelles and PTX-loaded micelles was evaluated in Caco-2 cells
164
using CCK-8 assay.22,23 The cells were seeded in 96-well plates at a density of 1 × 104 cells per well. After 2 days of
165
incubation, cells were incubated with 200 µL of test solutions diluted in fresh serum-free medium for 2 h at 37 ºC, using
166
200 µL of non-complete DMEM as negative control. The test solutions were divided into five groups: (1) DHC (0.1
167
mg/mL-1 mg/mL); (2) TGA-DHC (0.1 mg/mL-1 mg/mL); (3) Taxol® (PTX: 50 µg/mL-250 µg/mL); (4) PTX-DHC (PTX:
168
50 µg/mL-250 µg/mL); (5) PTX-TGA-DHC (PTX: 50 µg/mL-250 µg/mL). Subsequently, medium was removed, and 100
169
µL of serum-free DMEM with 10 µL of CCK-8 solution was added to the wells. The cells were then incubated for an
170
additional 0.5 h and the absorbance of samples was measured at 450 nm using a microplate reader (MultiskanMk3, Thermo
171
Labsystems, Beverly, MA, USA).
172 173 174
2.10. Uptake Studies. To evaluate the enhancement of cellular uptake of PTX offered by sulfhydrylated micelles and the related mechanism, the uptake studies were divided into three parts as follows. 6
ACS Paragon Plus Environment
Page 7 of 32
1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60
Molecular Pharmaceutics
175
2.10.1. Effect of PTX Concentration on Cell Uptake. Caco-2 cells were seeded in 24-well plates at a density of 1 × 105
176
cells per well. After 18-21 days of incubation, the cells were washed twice with Hank’s balanced salt solutions (HBSS) and
177
used for the uptake assays. 400 µL of Taxol® and PTX-loaded micelles diluted in DMEM (containing 50, 67, 100 µg/mL of
178
PTX) were added into each well. Subsequently, the test solutions were removed after culture at 37 ºC for 2 h and the cells
179
were washed three times with PBS at 4 ºC. The amount of PTX in Caco-2 cells was assayed by HPLC. Uptake ratio (U%)
180
was expressed as following:U% = QPTX in cells/Qcell protein× 100%; where QPTX in cells and Qcell protein represented the amount of
181
PTX in Caco-2 cells and cell protein respectively.
182
2.10.2. Endocytosis Pathways of PTX-TGA-DHC. Caco-2 cells were cultured with various endocytosis inhibitors to
183
demonstrate the pathways of cellular uptake. Herein, the cells were first incubated with 20 µg/mL of chlorpromazine
184
(inhibitor of clathrin-mediated endocytosis),24 15 µg/mL of nystatin (inhibitor of caveolin-mediated endocytosis)25 or 550
185
µM of amiloride (inhibitor of macropinocytosis)26. After incubation with the endocytosis inhibitors at 37 ºC for 1 h, the cells
186
were further cultured with PTX-TGA-DHC or PTX-DHC at a PTX concentration of 50 µg/mL. Finally, the test solutions
187
were removed and washed three times with PBS at 4 ºC. The amount of PTX in Caco-2 cells was assayed by HPLC.
188
2.10.3. Inhibition of P-gp Efflux Pumps by TGA-DHC Polymer. Caco-2 cells were cultured with different PTX
189
formulations (50 µg/mL of PTX) to evaluate the effect of TGA-DHC polymer on P-gp inhibition. After incubation with
190
either Taxol®, Taxol® with 100 µM of verapamil, Taxol® with 0.12 mg/mL of DHC polymer, Taxol® with 0.12 mg/mL of
191
TGA-DHC polymer, PTX-DHC, PTX-DHC with 100 µM of verapamil, PTX-TGA-DHC or PTX-TGA-DHC with 100 µM
192
of verapamil at 37 ºC for 2 h, the cells were washed with PBS three times at 4 ºC and the amount of cellular PTX was
193
assayed by HPLC.
194
High content cellular imaging system and flow cytometry were also employed to confirm the effect of TGA-DHC on
195
P-gp inhibition. R-123 loaded micelles (R-123-TGA-DHC, R-123-DHC) were prepared following the procedure for
196
PTX-TGA-DHC preparation except by replacing PTX with R-123. Caco-2 cells were seeded in 6-well plates at a density of
197
1 × 105 cells per well. The cells were washed twice with PBS before being incubated with free R-123 (5 µM), free R-123
198
with 100 µM of verapamil, R-123-DHC, R-123-DHC with 100 µM of verapamil, R-123-TGA-DHC, or R-123-TGA-DHC
199
with 100 µM of verapamil at 37 ºC for 2 h. Then, the cells were washed three times with 4 ºC PBS and observed by high
200
content cellular imaging system (ImageXpress, Molecular Devices, USA). Subsequently, cells were trypsinized and
201
collected through centrifugation for analysis by flow cytometry (BD FACSCantoTM, USA).
202 203
2.11. Transport Studies of PTX-TGA-DHC across Caco-2 Cells. Caco-2 cells were seeded at a density of 1 × 105 cells
204
per well in inserts containing permeable polycarbonate membrane (6.5 mm insert diameter, 3.0 µm pore size, 0.33 cm2
205
growth area) in 24-well plates. The cells were fed every two days for the first week, and then fed daily until used for 7
ACS Paragon Plus Environment
Molecular Pharmaceutics
1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60
Page 8 of 32
206
experiments 21 ~ 24 days after seeding. Transepithelial electrical resistance (TEER) values were measured with a Millicell®
207
electrical resistance system (Millipore, USA) to evaluate the integrity of the cell monolayers. ATEER value exceeding 350
208
Ω cm2 indicated a well-developed tight junction of the cells. In addition, the integrity of the cell monolayers was further
209
confirmed through Transmission electron microscopy (TEM, H-600, Hitachi, Japan).
210
Before the transport studies, the cell monolayers were washed twice with HBSS at 37 ºC for the apical (AP) and
211
basolateral (BL) side. Subsequently, the monolayers were pre-incubated with HBSS at 37 ºC for 15 min. To estimate the
212
transport from AP to BL side, 200 µL of test solutions was added to the AP side while 800 µL of blank HBSS was added to
213
the BL side. Test solutions included Taxol® (37 ºC), Taxol® with 100 µM of verapamil (37 ºC), PTX-DHC (37 ºC),
214
PTX-TGA-DHC (37 ºC), PTX-TGA-DHC (4 ºC), PTX-TGA-DHC with 20 µg/mL of chlorpromazine (37 ºC), and
215
PTX-TGA-DHC with 15 µg/mL of nystatin (37 ºC). Samples of 100 µL were taken from the receiving chamber and
216
supplemented with fresh HBSS at 0.5, 1, 1.5 and 2 h.Then samples were mixed with an equal volume of methyl cyanides,
217
vortexed, separated by centrifugation and analyzed by HPLC. Apparent permeability coefficients (Papp) of PTX could be
218
determined according to the equation:
219
Papp = (dQ/dt)/(A × C0),
220
where the dQ/dt (µg/s) was the drug permeation rate, A was the surface area of polycarbonate membrane (0.33 cm2) and C0
221
(µg/mL) was the initial concentration of PTX in the donor compartment at 0 h.
222 223
2.12. In Vitro Mucoadhesion Studies of TGA-DHC on Freshly Excised Rat Intestine. Rats were fasted for 12 hours
224
with water, anesthetized, and incised along the abdominal midline. The intestine was washed by pre-warmed saline at 37 ºC
225
until the effluent is clean. The intestinal segment of the rat was divided into four segments: duodenum, jejunum, ileum, and
226
colon. The different segments of the intestine were separated and 6 cm of each segment was obtained. Coumadin-6 loaded
227
micelles (C6-TGA-DHC, C6-DHC) were prepared following the procedure for PTX-TGA-DHC preparation except by
228
replacing PTX with coumadin-6. 0.4 mL of C6-TGA-DHC and C6-DHC diluted by pH 6.8 PBS was injected into the
229
separated intestinal segments with ligation on both sides and incubated at 37 ºC for 2 h. Micelles which were not absorbed
230
were removed by two washes with 1 mL of PBS. Tissue sections were stored in formalin and 7 µm thick slices were
231
obtained for florescent microscopy. In addition, after two washeswith PBS to remove the unabsorbed micelles, mucus layers
232
covering the treated intestinal segments were isolated by scraping off with a scalpel.27 The corresponding intestinal
233
segments minus the mucus layer and the obtained mucus samples were submerged into 4 mL of methanol and homogenized
234
by ultrasonication to dissolve coumadin-6 internalized within the intestine or detained in the mucus layer. After
235
centrifugation at 603 ×g for 10 min, the concentration of coumadin-6 in the supernatant was detected.
236 8
ACS Paragon Plus Environment
Page 9 of 32
1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60
Molecular Pharmaceutics
237 238
2.13. Statistical Analysis. All the experiments were repeated at least three times. Data were expressed as means ±
239
standard deviation (S.D.). All results were analyzed by 2-tailed Students t-test or one-way ANOVA, followed by the
240
Tukey-Kramer test. P < 0.05 was considered to be statistically significant.
241 242
3. RESULTS AND DISCUSSION
243
3.1. Synthesis and Characterization of Sulfhydrylated N-deoxycholic acid-N,O-hydroxyethyl Chitosan
244
(TGA-DHC). As shown in Scheme 2, the amphiphilic chitiosan-based conjugate, DHC, was synthesized and characterized
245
in our previous report.19 The DS of deoxycholic acid and hydroxyethyl in DHC determined by elemental analysis was 3.77%
246
and 106.57%, respectively. The objective sulfhydrylated micelle, TGA-DHC, was obtained through conjugating the
247
carboxyl of thioglycolic acid (TGA) to the free amino of DHC at different feeding ratios of reactants and various pH.
248
249 250
Scheme 2. Synthetic scheme of DHC and TGA-DHC conjugates.
251 252
The quantity of thiol in TGA-DHC was quantitatively determined by Ellman's method with or without NaBH4
253
pretreatment. Because thiol is susceptible to redox bylight, oxygen and metal ions into the disulfide form inner carriers,
254
reduced and oxidized thiol exist simultaneously in TGA-DHC.27 The quantity of thiol detected by Ellman's reagent without
255
pretreatment of NaBH4 represented the amount of free thiol, while those detected after reduction by NaBH4 represented both
256
the reduced and oxidized thiol groups (total amount). Results in Table 1 demonstrated that the total amount of thiol in
257
TGA-DHC increased as pH increased from 3 to 6. This trend may be attributed to the enhanced catalytic activity of EDC at
258
pH values close to, but less than 7.0. However, the pH-mediated change in the amount of free thiol in TGA-DHC was 9
ACS Paragon Plus Environment
Molecular Pharmaceutics
1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60
Page 10 of 32
259
inconsistent with the measured total thiol content. It was observed that the quantity of free thiol increased as pH increased
260
from 3 to 5. However, a subsequent decline in the amount of free thiol was observed in pH values above 5. This observation
261
can be primarily attributed to the formation of disulfide bonds from thiol in the oxidative environment.28 Thus generating
262
TGA-DHC with the maximal quantity of free thiol for mucosal adhesion enhancement could be obtained at pH 5.
263
Furthermore, results indicated that the grafting rate of thioglycolic acid was not improved when feeding ratio was increased
264
to 0.5:1 (n(DHC):n(TGA)) at pH of 5. Resultantly, the optimal TGA-DHC conjugate synthesized at pH 5 with the molar ratio
265
n(DHC):n(TGA):n(EDC):n(NHS) of 1:1:2:1 was used in the following article. Table 1. DS of thiol group measured by Ellman’s reagent of a series of TGA-DHC.
266 No. 1 2 3 4 5 6
n(DHC):n(TGA) :n(EDC):n(NHS) 1:1:2:1 1:1:2:1 1:1:2:1 0.5:1:2:1 2:1:2:1 1:1:2:1
The free amount of thiol (µmol/g) 65.5 137.3 286.8 203.7 193.9 145.8
pH 3 4 5 5 5 6
The total amount of thiol (µmol/g) 186.4 354.3 697.9 601.5 517.9 729.1
267 268
3.2. Preparation and Characterization of PTX-loaded TGA-DHC and DHC Micelles. PTX was loaded into DHC and
269
TGA-DHC micelles thorough sonication and dialysis. As shown in Table 2, PTX-DHC and PTX-TGA-DHC exhibited an
270
average size of 150 nm, a cationic charge of around +18 mV. An excellent PTX-loading capability was observed with a drug
271
loading (DL) content of ~30 wt% and a drug encapsulation efficiency (EE) of ~75%. Modification of TGA showed no
272
significant effect on the particle size, zeta potential or drug-loading ability of micelles (P > 0.05). TEM was used to
273
visualize the size and morphology of PTX-loaded micelles. Results in Figure 1A and Figure S1 validated that
274
PTX-TGA-DHC and PTX-DHC micelles displayed similar spherical morphology at approximately 100 nm diameter with a
275
narrow size distribution. The difference in size observed by TEM from those obtained from DLS was attributed to micellar
276
collapse during the drying processes of TEM sampling.
277 278
Table 2. Loading capacity of TGA-DHC and DHC micelles for PTX (n = 3).
PTX-DHC PTX-TGA-DHC
Diameter (nm)(µ2/Г2)
Zeta Potential (mV)
DL (wt%)
EE (%)
148.11 ± 5.42(0.116) 156.32 ± 6.93(0.098)
19.23 ± 3.11 17.83 ± 2.13
30.21 ± 1.83 31.91 ± 3.13
74.87 ± 3.43 79.67 ± 5.84
279 280
Furthermore, to investigate the physical state of PTX in micelles, WAXD and DSC analysis were performed for
281
PTX-TGA-DHC with PTX, TGA-DHC, and physical mixture of PTX + TGA-DHC as controls. The WAXD diagrams were
282
illustrated in Figure 1B. PTX showed intense peaks at 2θ of 5-15º and small peaks between 15 and 30º. Meanwhile, 10
ACS Paragon Plus Environment
Page 11 of 32
1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60
Molecular Pharmaceutics
283
TGA-DHC gave a broad peak at 15º ~ 30º. The typical crystal peaks of PTX and TGA-DHC were both present in the
284
WAXD diagram representing a physical mixture of PTX + TGA-DHC. On the other hand, the PTX-TGA-DHC micelles
285
possessed negligible characteristic peaks of PTX and exhibited peaks of free TGA-DHC. This finding confirms that input
286
PTX was encapsulated in micelles with reduced crystallinity. The results of DSC in Figure 1C further confirmed the
287
existing state of PTX in PTX-TGA-DHC. The PTX in the crystalline state showed an endothermic peak at 220.0 ºC and an
288
exothermic peak at 238.1 ºC, while TGA-DHC possessed a single, endothermic peak at 58.4 ºC. Additionally, the
289
representative peaks of PTX were seen for the physical mixture of PTX + TGA-DHC, but almost absent for
290
PTX-TGA-DHC micelles, indicating a notably reduction in the crystalline state of PTX in PTX-TGA-DHC.
291
When administered, orally delivered nanoparticles firstly interact with the strong acidic environments in gastric juice
292
before passing in to the mildly acidic environment in intestinal juice; finally passing into blood circulation. To simulate this
293
change, Taxol®, PTX-DHC micelles and PTX-TGA-DHC micelles were incubated with artificial gastric juice (pH 1.2) for 2
294
h followed by incubation with artificial intestinal fluid (pH 6.8) for another 22 h to simulate the PTX release profiles after
295
oral administration of micelles before transport across the epithelium. As shown in Figure 1D, both PTX-DHC and
296
PTX-TGA-DHC exhibited a slow rate of drug release. It was found that only 1.5% of PTX was released within 2 h after
297
administration in artificial gastric juice (pH 1.2) and only around 5.0% was released within 22 h in artificial intestinal fluid
298
(pH 6.8). These results indicated that PTX-DHC and PTX-TGA-DHC micelles retained structural integrity in the
299
gastrointestinal tract, with only minimal drug leakage. This property allows the benefit of increased oral bioavailability of
300
PTX delivered by PTX-TGA-DHC for transcytosis across enterocytes. Compared with the two micelles, a similar but
301
slightly faster drug release was observed from Taxol®. Specifically, around 12% of PTX was released after 24 h incubation
302
in the simulated condition. This result suggested improved oral absorption of PTX-DHC and PTX-TGA-DHC over Taxol®
303
not resulting from different release kinetics.
11
ACS Paragon Plus Environment
Molecular Pharmaceutics
1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60
Page 12 of 32
304 305
Figure 1. (A) TEM images of PTX-TGA-DHC micelles. XRD (B) and DSC (C) spectrum of blank TGA-DHC micelles, the
306
physical mixture of PTX + TGA-DHC, PTX-loaded TGA-DHC micelles and free PTX. (D) In vitro release of
307
PTX-TGA-DHC micelle, PTX-TGA micelles and Taxol®: 0-2 h in simulated gastric fluid (pH 1.2), 2-24 h in simulated
308
intestinal fluid (pH 6.8) (n = 6).
309 310
3.3. Pharmacokinetic Studies. To directly verify the advancement of PTX oral absorptive efficiency when delivered by
311
TGA-DHC, pharmacokinetic analysis of PTX-TGA-DHC after oral administration into rats at a PTX dosage of 20 mg/kg
312
was performed alongside orally delivered PTX-DHC and Taxol® as controls. Rats treated with a single i.v. administration of
313
Taxol® at 7 mg/kg were set as the reference group. The plasma PTX concentration plotted against time for each of these
314
four groups were shown in Figure 2. As exhibited in Figure 2A, the plasma concentration-time curves of the three oral
315
administrated formulations all represented a prominent hepatobiliary circulation-mediated bimodal effect. However, the
316
PTX plasma concentration decreased rapidly when treated with Taxol®, and reached non-detectable level after just 24 hours.
317
In contrast, PTX plasma concentrations when delivered using PTX-DHC or PTX-TGA-DHC remained above the 0.1 µg/mL
318
threshold even after 48 hours. Furthermore, the PTX-TGA-DHC group possessed a higher PTX plasma concentration at all 12
ACS Paragon Plus Environment
Page 13 of 32
1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60
Molecular Pharmaceutics
319
measured time points compared with PTX-DHC. The corresponding pharmacokinetic parameters of PTX in these
320
formulations were summarized in Table 3. As depicted, the oral absorption of PTX was extremely limited when
321
administrated as Taxol® with a bioavailability of 18.14% and AUC0-∞ of 13.99 ± 1.62 µg·h· mL-1. Oral administration of
322
PTX using DHC micelles resulted in a 1.3-fold increase in Cmax and a 2.5-fold increase in bioavailability compared with
323
Taxol®. The oral absorptive enhancement of this chitosan derivative micelle has been demonstrated and reported by our
324
group.19 However, the Cmax and bioavailability were further improved when PTX was orally delivered by TGA-DHC.
325
Specifically, PTX-TGA-DHC possessed a 3.2-fold greater AUC0-∞ and a 1.8-fold higher Cmax than Taxol®. TGA-DHC
326
micelles increased the bioavailability of PTX to 57.52%; 3.2-fold and 1.3-fold higher when compared with Taxol® and DHC
327
respectively. This result demonstrates that TGA-DHC was able to further promote the oral absorption of PTX over DHC.
328
Advantages presented herein may be attributed to a two faceted system composed of the sulfhydryl-mediated mucosal
329
adhesion and the enhanced oral PTX absorption by chitosan-based micelles. Firstly, improved mucosal adhesion allowed for
330
reduced clearance by mucus and a subsequently prolonged retention in the gastrointestinal tract. Secondly, chitosan-based
331
micelles help PTX absorbed through transcytosis cross the endothelium. Additionally, PTX delivered from chitosan
332
derivative polymers also benefit from P-gp inhibition.
333
334 335
Figure 2. (A) Mean pharmacokinetic profiles of PTX-DHC, PTX-TGA-DHC micelles and Taxol® after an oral dose of 20
336
mg/kg in rats (n = 3). (B) Mean pharmacokinetic profiles of Taxol® after an i.v. administration at a dose of 7 mg/kg (n = 3). 13
ACS Paragon Plus Environment
Molecular Pharmaceutics
1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60
Page 14 of 32
337 338
Table 3. Comparison of the mean pharmacokinetic parameters of PTX-DHC, PTX-TGA-DHC and Taxol® after a single oral
339
dose of 20 mg/kg in rats (n = 3). Parameter
Unit
Taxol® (7 mg/kg i.v.)
Taxol® (20 mg/kg p.o.)
PTX-DHC (20 mg/kg p.o.)
PTX-TGA-DHC (20 mg/kg p.o.)
Cmax AUC0-t
µg·mL-1 µg·h·mL-1
16.74 ± 1.66 17.89 ±1.85
0.76 ± 0.13 9.77 ± 1.03
1.00 ± 0.07* 24.39 ± 1.49**
1.36 ± 0.13**† 30.98 ± 2.21**†
AUC0-∞
µg·h·mL-1
18.85 ± 1.83
13.99 ± 1.62
30.53 ± 2.04**
42.87 ± 3.11**†
CL
mL·h-1
71.77 ± 6.51
245.17 ± 28.94
112.42 ± 4.80**
85.38 ± 11.03**
MRT
h
3.42 ± 0.24
22.32 ± 5.76
31.08 ± 7.89
37.77 ± 7.89*
F
%
N/A
18.14 ± 1.85
45.28 ± 2.52*
57.52 ± 3.51**†
340
*significantly different fromTaxol® (P < 0.05)
341
**significantly different from Taxol® (P < 0.01)
342
†
significantly different from PTX-DHC (P < 0.05)
343 344
3.3. Cellular Evaluation for the Mechanism of Oral Absorptive Enhancement by TGA-DHC. Having confirmed the
345
enhancement of oral bioavailability with PTX delivered using TGA-DHC, the mechanism of oral absorptive enhancement
346
was investigated using the Caco-2 cell line.
347
3.3.1. Determination of the Tested Concentration of PTX in Micelles in Cell Experiments. To ensure the feasibility and
348
accuracy of cellular experiments on Caco-2, the highest possible concentration of PTX delivered using micelles, or Taxol®
349
with negligible toxicity should be investigated. Firstly, the cytotoxicity of blank DHC micelles, TGA-DHC micelles,
350
PTX-loaded micelles and Taxol® on Caco-2 after 2 h of incubation was investigated through quantifying cell viability using
351
a cell counting kit-8 (CCK-8) assay. Results in Figure 3A indicated that DHC and TGA-DHC exhibited negligible toxicities
352
on Caco-2 cells even with a concentration up to 1 mg/mL, demonstrating the advantage of chitosan-based polymers in
353
biocompatibility. The relative cell viability of PTX-DHC, PTX-TGA-DHC and Taxol® group were all well above 90% when
354
the PTX concentrations were below 100 µg/mL, implying no significant toxicity toward Caco-2 cells. Yet, toxicities against
355
Caco-2 increased notably when the PTX concentration was raised to 250 µg/mL delivered using Taxol®. As a result,
356
PTX-DHC, PTX-TGA-DHC and Taxol® with the PTX concentrations of 100, 67, 50 µg/mL were used as the test
357
formulations in the following Caco-2 experiments to comparatively evaluate the effect of TGA-DHC micelles on enhancing
358
the oral absorption of PTX.
359
3.3.2. Uptake Studies in Caco-2 Cells. The ability of the DHC and TGA-DHC micelles to improve the cellular uptake of
360
PTX into Caco-2 was determined in comparison with Taxol®. Results in Figure 3C showed ~0.90 µg/mg PTX
361
accumulation was seen in Caco-2 cell when delivered by Taxol® at PTX concentrations of 50, 75, 100 µg/mL. In contrast, 14
ACS Paragon Plus Environment
Page 15 of 32
1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60
Molecular Pharmaceutics
362
Caco-2 cells treated with PTX-DHC at 50, 75, 100 µg/mL showed PTX concentrations of 12.85 ± 1.73, 13.36 ± 3.08 and
363
13.96 ± 2.64 µg/mg respectively. Meanwhile PTX-TGA-DHC treatment at the same concentrations of 50, 75, 100 µg/mL
364
displayed a PTX internalization amount of 11.66 ± 3.46, 14.01 ± 2.14 and 14.24 ± 3.04 µg/mg respectively. The ~15.5-fold
365
increase in uptake of PTX by Caco-2 cells treated by micelles over Taxol® demonstrated the appealing effects of
366
chitosan-based micelles on improving drug internalization. This finding may be explained by the encapsulation of drug
367
molecules into hydrophobic cores and transcytosis across cells in a manner that protects PTX from P-gp efflux. It was worth
368
noting that PTX accumulation concentrations were nearly identical in Caco-2 cells when delivered by TGA-DHC even
369
when the PTX incubation concentration increased to 100 µg/mL. Additionally, sulfhydrylation did not remarkably enhance
370
the drug intercellular delivery capability of chitosan-based micelles into Caco-2 cells. This finding may be due to the
371
deficiency of cysteine-rich mucosal glycoprotein around Caco-2 cells secreted by goblet cells in the intestinal epithelium.29
372 373
3.3.3. Endocytosis Pathways of PTX-TGA-DHC. It has been confirmed that TGA-DHC and DHC micelles were capable
374
of improving PTX accumulation in Caco-2 cells. Next, the cellular endocytosis-mediated enhancement offered by these two
375
micelles was investigated with regards to the mechanism of endocytosis. Uptake studies on Caco-2 were performed in the
376
presence of chlorpromazine, nystatin, or amiloride which inhibited clathrin-mediated endocytosis, caveolin-mediated
377
endocytosis, or macropinocytosis respectively. As depicted in Figure 3D, incubation with chlorpromazine resulted in a
378
significant decrease (P < 0.05) of cellular uptake of PTX into Caco-2 cells, while amiloride and nystatin had no significant
379
effect on the Caco-2 uptake of PTX. This finding indicates that PTX-DHC is permeated into cells mainly through the
380
clathrin-mediated endocytotic pathway. However, the uptake of PTX-TGA-DHC into Caco-2 cells was remarkably
381
decreased by chlorpromazine and nystatin (Figure 3E), implying that clathrin and caveolin were both involved in the
382
Caco-2 cellular internalization of PTX-TGA-DHC.
15
ACS Paragon Plus Environment
Molecular Pharmaceutics
1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60
Page 16 of 32
383 384
Figure 3. (A) Cytotoxicity of DHC and TGA-DHC against Caco-2 cells at different concentrations (n = 6). (B) Cytotoxicity
385
of Taxol®, PTX-DHC and PTX-TGA-DHC to Caco-2 cells (n = 6). (C) Cellular uptake of Taxol®, PTX-DHC and
386
PTX-TGA-DHC at PTX concentrations of 50, 67, 100 µg/mL into Caco-2 cells (n = 6, **P < 0.01 PTX-loaded micelles vs
387
Taxol® at the same PTX concentration). Relative uptake efficiency of PTX-loaded micelles with 50 µg/mL PTX in absence
388
or presence of various endocytosis inhibitors: (D) PTX-DHC, (E) PTX-TGA-DHC (n = 3, *P < 0.05 vs control, **P < 0.01
389
vs control). (F) Cell uptake of PTX into Caco-2 cells: Taxol®, Taxol® with verapamil, Taxol® with DHC, Taxol® with
390
TGA-DHC, PTX-DHC, PTX-TGA-DHC, PTX-DHC with verapamil and PTX-TGA-DHC with verapamil diluted at 50
391
µg/mL PTX (n = 3, *P < 0.05 vs Taxol®, **P < 0.01 vs Taxol®). (G) High content cellular imaging and flow cytometry
392
results of R-123 accumulation in Caco-2 cells: (a): free R-123, (b): free R-123 with verapamil, (c): free R-123 with
393
TGA-DHC, (d): R-123-TGA-DHC, (e) R-123-TGA-DHC with verapamil. Scale bar represents 100 µm. 16
ACS Paragon Plus Environment
Page 17 of 32
1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60
Molecular Pharmaceutics
394 395
As reported, chitosan-based micelles exhibited a profound effect on P-gp inhibition.17 To determine whether the
396
TGA-DHC and PTX-DHC micellar polymers could positively impact the cellular accumulation of PTX (a typical
397
P-gpsubstrate) through P-gp inhibition, an uptake experiment of Taxol®, PTX-DHC and PTX-TGA-DHC on Caco-2 cells
398
was further performed with or without the addition of verapamil, a traditional P-gp inhibitor. As shown in Figure 3F, the
399
cellular internalization of Taxol® (50 µg/mL PTX), in which PTX mainly exist as free molecules, could be significantly
400
increased (P < 0.05) through co-incubation with 100 mM of verapamil. When PTX-TGA-DHC or PTX-DHC was diluted to
401
the PTX concentration of 50 µg/mL, the concentration of micellar polymer was around 0.12 mg/mL. The presence of 0.12
402
mg/mL of TGA-DHC or DHC also resulted in a remarkably improved uptake of Taxol® (P < 0.05), demonstrating the P-gp
403
inhibitive effect of these two micellar polymers. However, the presence of verapamil did not significantly increase the
404
uptake efficiency of PTX-TGA-DHC or PTX-DHC micelles; likely due to the P-gp-independent cellular internalization of
405
the integrate micellar particles.
406
To further intuitively observe the improvement of TGA-DHC on the uptake of PTX, R-123 which is a substrate of P-gp,
407
was selected as fluorescence probe and loaded into micelles to evaluate the P-gp activity in Caco-2 cells. The intracellular
408
fluorescence intensity of R-123 in Caco-2 cells was tested in various combinations: cells incubated with 5 mM of free
409
R-123 solution with or without verapamil, cells incubated with free R-123 with TGA-DHC, and cells incubated with
410
R-123-TGA-DHC with or without 100 mM of verapamil. All groups were imaged using a high content cellular imaging
411
system. Furthermore, the intracellular fluorescence intensity of R-123 was quantitatively detected by flow cytometry. As
412
shown in Figure 3G, both verapamil and TGA-DHC increased the amount of internalized R-123 over the free R-123 group,
413
confirming that TGA-DHC possessed an inhibitive effect on P-gp efflux of R-123. Moreover, the intracellular fluorescence
414
intensity of R-123 was further enhanced when encapsulated and delivered by TGA-DHC. Meanwhile, the presence of
415
verapamil did not significantly increase the cellular accumulation of R-123-TGA-DHC. This phenomenon resulted from the
416
P-gp-independent internalization of R-123-TGA-DHC into Caco-2 cells, during which R-123 was loaded firmly in the inner
417
cores of TGA-DHC micelles and protected from the P-gp recognition. Resultantly, R-123-TGA-DHC effectively bypassed
418
the P-gp efflux. All results of this study suggest that TGA-DHC offers a promising delivery vehicle for P-gp substrates,
419
including pharmaceutical therapeutics, for oral administration.
420 421
3.3.4. Transport Studies of PTX-TGA-DHC. Despite the demonstration that TGA-DHC can effectively improve PTX
422
accumulation in Caco-2 cells, determination of the drug’s apparent permeability coefficient (Papp) is necessary owning that
423
PTX absorption through oral administration requires successful transport of PTX across intestinal epithelial cells into blood
424
circulation. Consequently, Caco-2 monolayers with the TEER higher than 350 Ω·cm2 were used to stimulate intestinal 17
ACS Paragon Plus Environment
Molecular Pharmaceutics
1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60
Page 18 of 32
425
epithelial cells and PTX Papp across the Caco-2 monolayers was then detected.5,30 Herein, a higher Papp value represented
426
greater oral absorption efficiency. As shown in Figure 4A, the TEER of the Caco-2 cell monolayer was above 350 Ω·cm2
427
after 21 days of cultivation, indicating well-developed tight junctions between cells. This tight junction in Caco-2 cell
428
monolayers was also intuitively demonstrated by TEM images in Figure 4B. In this study, Caco-2 monolayers were treated
429
with PTX-TGA-DHC (50 µg/mL PTX), PTX-DHC (50 µg/mL PTX), Taxol® (50 µg/mL PTX) with or without 100 mM
430
verapamil, and Taxol® (50 µg/mL PTX) with 0.12 mg/mL TGA-DHC polymer to comparatively investigate the ability of
431
TGA-DHC to enhance PTX transport (Figure 4C). Consistent with previous reports, the Papp of Taxol® was low, ranging
432
around 2.13 ± 0.22 × 10-6 cm/s despite the presence of Cremophor EL; a surfactant speculated to inhibit P-gp efflux
433
pumps.31 Such a considerably low Papp of Taxol® was attributed to P-gp efflux as PTX existed as free molecules in the
434
formulation system of Taxol®. Thus, a poor oral absorption of Taxol® was observed, which has been illustrated in
435
pharmacokinetic studies. Yet, it was exhibited that 100 mM of verapamil significantly improved the Papp of Taxol® (P
0.05). Collectively, clathrin was implicated in both
463
the cellular endocytosis and intestinal transport mechanisms of PTX-TGA-DHC, whereas caveola was only associated with
464
the cellular endocytosis but not with the intestinal transcytosis.
465 466
Figure 4. (A) TEER of Caco-2 cell monolayersover 22 days (n = 3). (B) Electron microscopy of Caco-2 cell monolayers. (C)
467
Apparent permeability coefficients of PTX: Taxol®, Taxol® with verapamil, Taxol® with TGA-DHC, PTX-DHC and
468
PTX-TGA-DHC micelles diluted to 50 µg/mL PTX at 37 ºC (n = 3, *P < 0.05 vs Taxol® at 37 ºC, **P < 0.01 vs Taxol® at
469
37 ºC). (D) TEER at the beginning and end of PTX-DHC and PTX-TGA-DHC transport experiments (n = 3). (E) Apparent
470
permeability coefficients of PTX for PTX-TGA-DHC at 37 ºC, PTX-TGA-DHC at 4 ºC, PTX-TGA-DHC with
471
chlorpromazine at 37 ºC and PTX-TGA-DHC with nystatin at 37 ºC (n = 3, *P < 0.05 vs PTX-TGA-DHC at 37 ºC, **P