46
Accepted Manuscript Title: Alternation of adriamycin penetration kinetics in MCF-7 cells from 2D to 3D culture based on P-gp expression through the Chk2/p53/NF-B pathway Author: Meng Lu Fang Zhou Kun Hao Jiali Liu Qianying Chen Ping Ni Honghao Zhou Guangji Wang Jingwei Zhang PII: S0006-2952(14)00691-1 DOI: http://dx.doi.org/doi:10.1016/j.bcp.2014.11.010 Reference: BCP 12141 To appear in: BCP Received date: 20-9-2014 Revised date: 17-11-2014 Accepted date: 19-11-2014 Please cite this article as: Lu M, Zhou F, Hao K, Liu J, Chen Q, Ni P, Zhou H, Wang G, Zhang J, Alternation of adriamycin penetration kinetics in MCF-7 cells from 2D to 3D culture based on P-gp expression through the Chk2/p53/NF-rmkappaB pathway, Biochemical Pharmacology (2014), http://dx.doi.org/10.1016/j.bcp.2014.11.010 This is a PDF file of an unedited manuscript that has been accepted for publication. As a service to our customers we are providing this early version of the manuscript. The manuscript will undergo copyediting, typesetting, and review of the resulting proof before it is published in its final form. Please note that during the production process errors may be discovered which could affect the content, and all legal disclaimers that apply to the journal pertain.

Alternation of adriamycin penetration kinetics in MCF-7 cells from 2D to 3D culture based on P-gp expression through the Chk2/p53/NF-B pathway

  • Upload
    gucas

  • View
    0

  • Download
    0

Embed Size (px)

Citation preview

Accepted Manuscript

Title: Alternation of adriamycin penetration kinetics in MCF-7cells from 2D to 3D culture based on P-gp expression throughthe Chk2/p53/NF-�B pathway

Author: Meng Lu Fang Zhou Kun Hao Jiali Liu QianyingChen Ping Ni Honghao Zhou Guangji Wang Jingwei Zhang

PII: S0006-2952(14)00691-1DOI: http://dx.doi.org/doi:10.1016/j.bcp.2014.11.010Reference: BCP 12141

To appear in: BCP

Received date: 20-9-2014Revised date: 17-11-2014Accepted date: 19-11-2014

Please cite this article as: Lu M, Zhou F, Hao K, Liu J, Chen Q, Ni P, Zhou H, WangG, Zhang J, Alternation of adriamycin penetration kinetics in MCF-7 cells from 2D to3D culture based on P-gp expression through the Chk2/p53/NF-rmkappaB pathway,Biochemical Pharmacology (2014), http://dx.doi.org/10.1016/j.bcp.2014.11.010

This is a PDF file of an unedited manuscript that has been accepted for publication.As a service to our customers we are providing this early version of the manuscript.The manuscript will undergo copyediting, typesetting, and review of the resulting proofbefore it is published in its final form. Please note that during the production processerrors may be discovered which could affect the content, and all legal disclaimers thatapply to the journal pertain.

Page 1 of 45

Accep

ted

Man

uscr

ipt

1

Title page

Title

Alternation of adriamycin penetration kinetics in MCF-7 cells from 2D to 3D culture

based on P-gp expression through the Chk2/p53/NF-κB pathway

Author names and affiliations

Meng Lu a,1, Fang Zhou a,b,1, Kun Hao a,b, Jiali Liu a, Qianying Chen a, Ping Ni a,

Honghao Zhou c, Guangji Wang a,b,* and Jingwei Zhang a,b,**

a Key Lab of Drug Metabolism and Pharmacokinetics, State Key Laboratory of

Natural Medicines, China Pharmaceutical University, Nanjing, Jiangsu, China.

b Jiangsu Key laboratory of drug design and optimization, China Pharmaceutical

University, Nanjing, Jiangsu, China.

c Institute of Clinical Pharmacology, Central South University, Changsha, Hunan,

China.

1 M. Lu and F. Zhou contributed equally to this work.

* Corresponding author: Professor Guangji Wang, Key Lab of Drug Metabolism and

Pharmacokinetics, China Pharmaceutical University, 24 Tong Jia Xiang, Nanjing,

Jiangsu, 210009, China. Tel: 86-25-83271128, Fax: 86-25-83271060, E-mail:

[email protected]

** Corresponding author: Associate Professor Jingwei Zhang, Key Lab of Drug

Metabolism and Pharmacokinetics, China Pharmaceutical University, 24 Tong Jia

Xiang, Nanjing, Jiangsu, 210009, China. Tel: 86-25-83271176, Fax: 86-25-83271060,

E-mail: [email protected]

Page 2 of 45

Accep

ted

Man

uscr

ipt

2

Authorship contribution statement

(1) Study conception and design;

G.J. Wang, F. Zhou, H.H. Zhou and J.W. Zhang

(2) Acquisition, analysis and/or interpretation of data;

M. Lu, J.L. Liu, Q.Y. Chen and P. Ni

(3) Drafting/revision of the work for intellectual content and context;

M. Lu, J.W. Zhang and K. Hao

(4) Final approval and overall responsibility for the published work.

G.J. Wang, F. Zhou and J.W. Zhang

Page 3 of 45

Accep

ted

Man

uscr

ipt

3

Abstract

Monolayer cells are largely different from tumor masses, and might misguide drug

screenings. 3D in vitro cell culture models simulate the characteristics of tumor

masses in vivo and have recently been used in many studies of anti-cancer drugs.

Among various 3D cell culture models, multi-cellular layer (MCL) models allow for

the direct quantitative assessment of the penetration of chemotherapeutic agents

through solid tissue environments without requiring the use of fluorescently labeled

drugs or imaging molecules. Therefore, in our present study, a 3D-no base and

embedded MCF-7 MCL model was successfully developed for a 14-day culture. Over

time, its thickness and cell layers increased and exhibited highly proliferative

properties and drug resistance to adriamycin (ADR) with markedly elevated IC50

values. Meanwhile, G2/M stage cycle arrest was also observed, which likely

up-regulated P-gp expression through the Chk2/p53/NF-κB pathway. The elevated

P-gp expression altered the ADR penetration kinetics in MCF-7 MCLs in vitro by

accelerating the apparent penetration of ADR through the intercellular spaces of the

MCLs. Additionally, a decreased ADR retention within tumor cells was observed, but

could be significantly reversed by a P-gp inhibitor. The attenuated ADR retention in

the deeper cells of tumor masses was confirmed in xenografted mice in vivo. This

phenomenon could be elucidated by the mathematical modeling of penetration

kinetics parameters. Our study provided a new model that evaluated and improved the

quantification of the drug penetration kinetics, revealed the relationship between P-gp

and drug penetration through tumor masses, and suggested the potential molecular

mechanisms.

Keywords

Multi-cellular layer; Penetration kinetics; P-gp; Chk2/p53/NF-κB pathway

Page 4 of 45

Accep

ted

Man

uscr

ipt

4

Chemical compounds studied in this article:

Adriamycin (PubChem CID: 31703); LY335979 (PubChem CID: 153997); AZD7762

(PubChem CID: 11152667); Nutlin-3 (PubChem CID: 216345); DMSO (PubChem

CID: 679); Propidium iodide (PubChem CID: 104981); MTT formazan (PubChem

CID: 16218671)

Page 5 of 45

Accep

ted

Man

uscr

ipt

5

Abbreviations

ADR (adriamycin); AUC (area under the concentration-time curve); ECM,

(extracellular matrix); ESI (electrospray ionization); HBSS (Hank’s balanced salt

solution); LC-MS/MS (liquid chromatography tandem mass spectrometry); MCL

(multi-cellular layer); MCR (multi-cellular resistance); MCS (multi-cellular spheroid);

MDR (multi-drug resistance); MRM (multiple reaction monitoring); P-gp

(P-glycoprotein); PI (propidium iodide); qPCR (quantitative real-time PCR); S.E.

(standard error).

Page 6 of 45

Accep

ted

Man

uscr

ipt

6

1. Introduction

In order to achieve therapeutic efficacy, sufficient concentrations of anti-cancer drugs

should thoroughly penetrate tumor masses to gain full access to all viable cancer cells.

However, most conventional anti-cancer drugs are confined to only the periphery of

tumor masses near the vasculature [1, 2], a phenomenon that is known as

multi-cellular resistance (MCR) [3, 4]. This is one of the main reasons for treatment

failure of anti-cancer drugs. The many mechanisms responsible for MCR are

interrelated and multi-faceted [5, 6] and include the over-expression of efflux pumps

through cytokine alternations [7] and cell cycle changes [8]. Therefore, new

anti-cancer agents that account for the spatial structure of tumor masses should be

developed. However, in vitro cultures of cell monolayers are still the most common

choice in conventional studies, and these differ from physiological tumor masses. The

majority of anti-cancer agents has been tested on cell monolayers and may thus

provide results and interpretations that are not applicable for clinical use.

Due to the non-physiological environments represented by monolayer cell

cultures, various 3D cell culture models have been developed for more accurate drug

evaluations. Multi-cellular layer (MCL) and multi-cellular spheroid (MCS) systems

are two important models that can reproduce the characteristics of tumor masses in

vivo and have been used widely in many pharmacokinetic and pharmacodynamic

studies of anti-cancer drugs [9, 10]. MCS systems are believed to more accurately

represent the structures and biochemical properties of tumor masses than do MCL

systems [11, 12]. However, for kinetics analyses, MCS systems have their limitations.

Accurate drug concentrations cannot be determined at different depth within a cell

spheroid or can only be achieved using semi-quantitative fluorescence-based imaging

[13-15]. MCL models provide for the direct quantitative assessment of the penetration

Page 7 of 45

Accep

ted

Man

uscr

ipt

7

of chemotherapeutic agents through solid tissue environments without the need for

fluorescent labeling. Thus, MCL models can be used to obtain the necessary

information on drug penetration and distribution properties through multi-cell barriers

for optimizing drug delivery [16-18].

In our previous studies, we analyzed the cellular pharmacokinetic mechanisms

of multi-drug resistance (MDR) of breast cancer cell MCF-7 monolayers induced by

the anti-cancer agent, adriamycin (ADR). Additionally, we screened for an effective

MDR reversal agent, ginsenoside Rh2 [19]. However, because breast cancer is a solid

tumor and presents MCR, there is a pressing need to analyze the penetration kinetics

of anti-cancer agents and explore strategies to optimize tumor tissue penetration.

Therefore, MCF-7 MCL models were developed in our present study. The penetration

kinetics of ADR on MCF-7 monolayers and MCL models were investigated,

compared and mathematically modeled. Xenograft in vivo tumor models were also

performed for confirmation. Finally, the MCR of MCF-7 MCL models and its

pharmacokinetic-related mechanisms were tentatively elucidated.

2. Materials and Methods

2.1 Materials

Adriamycin (purity > 99%) was purchased from Shenglin Chemical industry (Jiangsu,

China). LY335979 and AZD7762 were purchased from Selleckchem (Houston, Texas,

USA). 3-(4,5-Dimethyl-2-thiazolyl)-2,5-diphenyl-2H-tetrazolium bromide (MTT),

propidium iodide (PI), Nutlin-3, DMSO, HPLC-grade methanol and formic acid were

purchased from Sigma Chemical Co. (St. Louis, MO, USA). Deionized water was

prepared by a Milli-Q system (Merck Millipore, Billerica, MA, USA) and was used

throughout. Matrigel, FITC-conjugated anti-P-gp antibody and isotype antibody were

Page 8 of 45

Accep

ted

Man

uscr

ipt

8

purchased from BD Biosciences (Franklin Lakes, New Jersey, USA). Standard cell

culture inserts (CM 6.5 mm, pore size 0.4 μm) were purchased from Merck Millipore

(Billerica, MA, USA). Monoclonal antibodies against Chk2, p-Chk2, p53, p65, Lamin

B, and horseradish peroxidase-conjugated goat anti-mouse/rabbit IgG secondary

antibodies were purchased from Cell Signaling Technology (Danvers, MA, USA).

The antibody for GAPDH was purchased from Bioworld Technology (Dublin, Ohio,

USA). The SYBR Prime Script RT-PCR Kit was purchased from Takara Bio Inc.

(Otsu, Shiga, Japan).

2.2 Animal welfare and Ethical statements

Healthy female BALB/c nude mice (18-22 g and 8-10 weeks old) were obtained from

Shanghai SLAC Laboratory Animal Co., Ltd. (Shanghai, China), and were kept ten

per cage at room temperature (22 ± 1 °C) with 50-60% relative humidity and

automatic day-night rhythm (12 h-cycle) in an SPF-grade environment. Tumors were

generated by subcutaneous injections of 5×106 exponentially growing MCF-7 cells

into the right flank regions of female athymic nude mice. Estrogen pellets were

implanted into the nude mice one day before injection of cells. Prior to each

experiment, the animals were fasted overnight (12 h) with free access to water. All

animal experiments were approved by the Animal Ethics Committee of China

Pharmaceutical University (Nanjing, Jiangsu, China). This study was carried out in

strict accordance with the Guidelines for Animal Experimentation of this institution.

All procedures were as humane as possible. Every effort was made to minimize

animal pain, suffering and distress and to reduce the number of animals used.

2.3 Monolayer Cell Culture

Page 9 of 45

Accep

ted

Man

uscr

ipt

9

MCF-7 human breast carcinoma cells were purchased from American Type Culture

Collection. Cells were grown in monolayers using RPMI 1640 medium supplemented

with 10% fetal bovine serum, and 100 U·ml-1 penicillin and streptomycin (Life

Technologies, Carlsbad, CA, USA). The cells were grown in an atmosphere of 5%

CO2 at 37 °C, and cell medium was changed every other day.

2.4 Multi-cellular Layer Culture

The multi-cellular layer (MCL) culture of MCF-7 cells was developed according to a

“3D-no base and embedded” model with minor modifications [20]. Briefly, cells were

suspended in serum-free medium containing 6% matrigel and seeded onto the

uncoated cell culture inserts. After incubating at 37 °C for 1 h to ensure matrigel

solidification, cell culture media was added. The cells were further incubated at 37 °C

to form MCLs; subsequently, the medium was changed daily.

2.5 Histology Assays and Packing density Measurements

Cells were fixed in 10% neutral-buffered formalin for 24 h, processed with gradient

concentrations of ethanol, placed in xylene overnight and subsequently, embedded in

paraffin. Sections were cut at a thickness of 4 μm and stained with H&E and Ki-67.

The thickness of the MCL was determined with a microscope (Leica, Wetzlar,

Germany). The packing density was calculated as the percentage of nuclear areas in

all areas of MCF-7 cells. The number of nuclei per unit surface area in each image

was quantified using Leica Qwin Lite.

2.6 Cell Cycle Analysis

Page 10 of 45

Accep

ted

Man

uscr

ipt

10

The cell cycle distribution was assayed by determining the DNA content of MCF-7

cells. Cells were fixed in 70% ethanol overnight at 4 °C, and re-suspended in a

staining solution containing RNase A and propidium iodide (PI) for 30 min. After

washing, the DNA content was determined by flow cytometry (FACS Calibur, BD,

Franklin Lakes, New Jersey, USA) and analyzed with CELLQUEST software.

2.7 Cytotoxicity Assay

MCF-7 cells were exposed to various concentrations of ADR for 72 h at 37 °C with

5% CO2. The cell sensitivities were determined by measuring cell growth inhibition

via MTT colorimetric assay. The IC50 values were calculated from survival curves

using the Bliss method.

2.8 Quantitative Real-time PCR Assay

The quantitative real-time PCR (qPCR) reactions were performed in a CFX96

Real-Time PCR Detection System (Bio-Rad, Hercules, CA, USA) using the SYBR

Premix Ex Taq II (Takara, Otsu, Shiga, Japan). The primers were synthesized by

Invitrogen (Life Technologies, Carlsbad, CA, USA) and the sequences of mdr1, bcrp,

mrp2 and reference gene β-actin were as follows:

mdr1: forward primer: GCTGGGAAGATCGCTACTGA;

reverse primer: GGTACCTGCAAACTCTGAGCA

bcrp: forward primer:AGATGGGTTTCCAAGCGTTCAT;

reverse primer: CCAGTCCCAGTACGACTGTGACA

mrp2: forward primer: ACAGAGGCTGGTGGCAACC;

reverse primer: ACCATTACCTTGTCACTGTCCATGA

β-actin: forward primer: GCGTGACATTAAGGAGAAG;

Page 11 of 45

Accep

ted

Man

uscr

ipt

11

reverse primer: GAAGGAAGGCTGGAAGAG.

qPCR conditions were 95 °C for 60 s, followed by 40 cycles of 95 °C for 5 s, 60 °C

for 30 s, and 72 °C for 30 s. The specificity of the primer was monitored using

product melting curves in each reaction well. The relative gene expressions of the

examined genes were normalized to the expression of the housekeeping gene β-actin.

2.9 P-gp Expression Assay

Cells were fixed with 4% paraformaldehyde solution, followed by washing and

blocking. Then, the cells were incubated with the FITC-conjugated anti-P-gp

polyclonal antibody or the isotype-matched negative control for 1 h at 37 °C. After

washing, cells were analyzed by flow cytometry for P-gp protein expression.

2.10 Cellular Retention Assay

Briefly, cells were incubated with Hank's Balanced Salt Solution (HBSS; 37 °C, pH

7.4) containing ADR for 2 h. The accumulation was stopped by rinsing the cells with

ice-cold HBSS. Cells were then lysed and protein concentrations were measured by

the Bradford method using the BCA protein assay kit (Beyotime, Jiangsu, China). The

ADR concentration was determined by LC-MS/MS as previously described [19]. All

experiments were conducted in triplicate.

2.11 Drug Penetration Assay

MCF-7 monolayers and MCLs were seeded on cell culture inserts and cultured for

designated times. Then, the cultures were incubated with HBSS (37 °C, pH 7.4)

containing ADR on the top chambers, and samples were collected from the bottom

chambers at each time point. At the end of the experiment, cells attached to the insert

Page 12 of 45

Accep

ted

Man

uscr

ipt

12

membranes were also collected. The concentration of ADR was determined by

LC-MS/MS as previously described, and protein concentrations were measured by the

Bradford method. All experiments were conducted in triplicate.

2.12 Mathematical Model of Drug Penetration Kinetics

ADR penetration kinetics was modeled using a one-compartment absorption model. A

brief schematic presentation of this model is shown in Fig 1, where c1, c2, and c3 are

the concentrations of ADR in the top chamber, the multi-cellular layer and the bottom

chamber, respectively, and ka and ke are the absorption rate and elimination rate

constants. An integrated mathematical model was constructed as differential functions

in Equations (1) - (4). All parameters were estimated by ADAPT 5 software [21].

2.13 Distribution of ADR in tumors in vivo

Mice bearing MCF-7 subcutaneous tumors in right flank regions were divided

randomly into two groups (ten mice per group). One group was pre-treated with

normal saline (i.v.) followed by ADR (30 mg·kg-1, i.v.), and the other group was

pre-treated with LY335979 (25 mg·kg-1, i.v.) followed by ADR (30 mg·kg-1, i.v.).

Three hours after ADR administration, blood samples were collected via the

Page 13 of 45

Accep

ted

Man

uscr

ipt

13

retro-orbital venous sinus into heparinized tubes, mice were sacrificed by CO2

asphyxiation and cervical dislocation, and tumors were collected. The tumors were

immediately embedded in OCT compound (Sakura, Torrance, CA, USA), frozen and

stored at -80 °C. Cryosections 10 μm-thick were cut at approximately 100 μm

intervals for each tumor and mounted on glass slides.

The red fluorescence of ADR was imaged and quantified with a confocal laser

scanning microscope (Axiovert 200M, Carl Zeiss, Oberkochen, Germany). The

fluorescent intensities of ADR in tumor cryostat sections were plotted against the

distance from the periphery region (set as 0) to the central region.

2.14 Western Blotting Assay

For Western blots, nuclear, cytoplasmic and whole cell extracts were prepared as

previously described [22]. The protein samples were separated on a 10%

SDS-polyacrylamide gel and transferred onto a polyvinylidene difluoride membrane

(Bio-Rad, Hercules, CA, USA). After blocking with 5% non-fat milk, the membrane

was incubated with the primary antibodies overnight at 4 °C, followed by horseradish

peroxidase-conjugated secondary antibody for 1 h at 37 °C. The signals were detected

using an enhanced chemiluminescence kit (Thermo Fisher Scientific, Waltham, MA,

USA) and was captured using a ChemiDoc XRS+ System (Bio-Rad, Hercules, CA,

USA).

2.15 Statistical Analysis

All the data are presented as means ± S.E. of at least three independent experiments.

The statistical analyses included two-tailed Student’s t-tests and one-way analysis of

Page 14 of 45

Accep

ted

Man

uscr

ipt

14

variances. Differences were considered significant at * p<0.05, ** p<0.01, or ***

p<0.001.

3. Results

3.1 Morphology and proliferative features of MCF-7 MCLs during the culture

time

As seen in Fig 2A, the structures of the MCF-7 MCL were compact and no regions of

necrosis were found. The thickness and cell layers of the MCL increased over the

culture time, but the packing density did not significantly change (Table 1).

Furthermore, all cells within the MCF-7 MCL exhibited vigorous proliferation as

evidenced by the strong Ki-67 staining (Fig 2B). However, the cell cycle analyses

indicated a significant cell cycle arrest at the G2/M phase (Fig 2C). Relative to the 3

days group, the percentage of cells in the G2/M phase increased by 1.43-, 1.76-, 1.98-

and 2.18-fold in a time-dependent manner (Fig 2D).

3.2 ADR resistance during the growth of MCF-7 MCL

The MCF-7 monolayer cells were sensitive to ADR with an IC50 at 0.75 μM (Fig 3A).

In the earlier culture times of the MCL (3 and 5 days), the IC50 of MCF-7 cells ranged

from 0.7 to 1 μM and were not significantly different from those of the monolayer

cells (Fig 3B and 3C). However, the IC50 values markedly increased with the culture

time at 8, 10 and 14 days by 15.8-, 18.28-, and 24.11-fold, respectively, compared

with those of the monolayer cells (Fig 3D-F).

3.3 Up-regulated gene and protein expressions of P-gp during the growth of

MCL

Page 15 of 45

Accep

ted

Man

uscr

ipt

15

As shown in Fig 4A, there were no significant changes for the gene expressions of

bcrp and mrp2 throughout the entire culture time. However, the mdr1 gene expression

was increased in a time-dependent manner during the culture time of the MCF-7 MCL,

especially on the 10th and 14th day of the MCL culture when approximately 110-fold

increases were observed compared with those of cell monolayers. Furthermore, the

mdr1 protein levels were also markedly elevated (Fig 4B), with an approximately

1.95-fold increase in the P-gp protein level on the 14th day of the MCL culture

compared with that of the MCF-7 monolayer (Fig 4C).

3.4 Decreased intracellular retention of ADR in MCF-7 MCL and its reversal by

P-gp inhibitor

As shown in Fig 4D, the intracellular retentions of ADR in the MCF-7 MCL were

significantly decreased in a time-dependent manner compared with that in monolayer

cells. On the 10th and 14th day of the MCL culture, there were decreases of

approximately 35.4% and 40.2% compared with those in monolayer cells. These

decreases were significantly reversed by treating with the P-gp specific inhibitor

LY335979, resulting in 1.44- and 1.76-fold increases, respectively.

3.5 Chk2/p53/NF-κB-mediated P-gp up-regulation in MCL culture

As seen in Fig 5A, the expressions of p-Chk2, Chk2, p53 and p65 were elevated in a

time-dependent manner in the MCF-7 MCL. Especially on the 14th days of the MCL

culture, the expressions of p-Chk2, p53, and nuclear p65 increased by 348%, 227%

and 194%, respectively (Fig 5B). When checkpoint kinase inhibitor AZD7762 was

added to the MCF-7 MCL that was cultured for 14 days, the up-regulated expressions

of p-Chk2 and Chk2 were significantly lowered in concentration-dependent manners

Page 16 of 45

Accep

ted

Man

uscr

ipt

16

(Fig 5C). The expressions of p53, p65, and P-gp were also down-regulated (Fig 5D

and 5E). When p53 activator Nutlin-3 was added in combination with AZD7762 in

the MCF-7 MCL, the down-regulations of p53 and p65 caused by AZD7762 were

reversed (Fig 5F).

3.6 Altered ADR penetration through MCF-7 MCL and its reversal by P-gp

inhibitor

As shown in Fig 6A, the P-gp inhibitor LY335979 exhibited no effects on the

penetration of ADR in the MCF-7 monolayer. When the MCF-7 MCL was cultured

for 10 days, while the amount of ADR penetrating to the bottom chamber of the cell

culture insert did not change compared with that of the MCF-7 monolayer, it was

significantly decreased when the culture was treated with LY335979 (Fig 6B).

However, the amount of ADR penetrating the MCF-7 MCL that was cultured for 14

days was significantly greater than that of the monolayer (by approximately 4-fold).

Again, this was also markedly decreased by LY335979 (Fig 6C). The intracellular

retentions of ADR within the cells constituting the monolayer or the MCL were

sharply decreased over time to form the MCL (from 8.15 ± 0.96 to 2.06 ± 0.31 and

0.47 ± 0.06). This decrease was markedly reversed by the P-gp inhibitor (Fig 6D).

This was also demonstrated by the fluorescent imaging of the cryosections of these

cells attached to the cell culture insert (Fig 6E). The plot of fluorescent intensities

against the distance from the top of the cell culture insert revealed that much more

ADR accumulated in the bottom cells in the presence of the P-gp inhibitor. This

promoted the penetration of ADR into the deeper cells of the MCL (Fig 6F). The

AUC of this intensity-distance curve increased by 1.7-fold in the presence of the P-gp

inhibitor (Fig 6G).

Page 17 of 45

Accep

ted

Man

uscr

ipt

17

3.7 Altered kinetics parameters of ADR penetration through MCF-7 MCL due to

P-gp inhibitor

The mathematical model reasonably described the penetration kinetics of ADR

through the MCF-7 MCL with or without LY335979. As shown in Fig 6H-J, the

fitting results matched reasonably well with the measured values. As seen in Table 2,

when ADR treatment was combined with LY335979, the maximum additive value of

ke (ke-add) and the maximum inductive value of ke (ke-induct) were significantly lowered.

3.8 ADR penetration in MCF-7 xenografts

When MCF-7 xenografted nude mice were administered ADR (30 mg·kg-1, i.v.) in the

presence of the P-gp inhibitor, the drug concentrations in plasma (Fig 7A) and tumor

(Fig 7B) were significantly increased by 1.5- and 1.8-fold, respectively. The mean

diameter of the MCF-7 xenograft was 1.2 mm. Its cryosections were divided into

three concentric regions (Fig 7C; the edge was set as a reference): the peripheral

region (0-200 μm), the intermediate region (200-400 μm) and the central region

(400-600 μm). The representative images for each region showed that ADR primarily

accumulated at the peripheral region of the xenograft with a decreasing concentration

toward the center. Low ADR fluorescence signals were observed in the intermediate

and central regions. In the presence of the P-gp inhibitor, the fluorescence intensities

of all three regions were markedly augmented, especially for the intermediate and

central regions (Fig 7D). As shown in Fig 7E, the fluorescence intensity against the

distance from the periphery to center of the xenograft was plotted. The whole curve

was up-shifted in the presence of the P-gp inhibitor with a 2.09-fold AUC increase

(Fig 7F).

Page 18 of 45

Accep

ted

Man

uscr

ipt

18

4. Discussion and conclusions

Most studies on the resistance of anti-cancer drugs have focused on molecular

mechanisms within individual cells [23, 24]. However, the vast physiological

differences between a single cell in vitro and a tumor mass in vivo often leads to the

failure of anti-cancer drug from in vitro screening to in vivo demonstration [25-27]. A

tumor mass contains not only various single cells, but also a microenvironment

around each cell and the 3D structure of cell clusters [28]. Therefore, it is necessary to

apply a 3D cell model to evaluate new drugs and their mechanisms [3, 29, 30].

Many papers have described the MCL culture of colorectal carcinoma cells [16,

17, 31]. To the best of our knowledge, this present study is the first time breast cancer

cells MCF-7 have been cultured as MCLs using a 3D-no base and embedded model.

Hence, various cell seeding densities, cell embedding methods, and different cell

insert materials were tested and compared (data not shown). A 3D-no base and

embedded MCF-7 MCL 14-day culture was successfully developed, containing 10-12

cell layers at a 40 μm thickness (Table 1). All cells constituting the MCLs were viable

and proliferative as indicated by strong H&E and Ki-67 staining, respectively (Fig 2A

and 2B). Cell cycle analyses also indicated that most cells were at the G1 phase,

followed by the S and G2/M phases, which are common observations for cancer cells

without drug treatment (Fig 2C). Because many previous studies have reported that

3D cell cultures exhibit native resistances to chemo-therapy drugs [32, 33], the

sensitivities of the MCF-7 MCLs were evaluated using ADR as the probe drug. The

IC50 values of ADR were increased by 25-fold compared with those of 2D cells (Fig

3); MCF-7 MCLs indeed became MCR. Thus, our established MCF-7 MCL model

was credible.

Page 19 of 45

Accep

ted

Man

uscr

ipt

19

Next, we tried to find out the mechanisms that lead to MCR during the 2D to

3D transitioning culture of MCF-7 cells. It is well-known that efflux pumps are the

main causes to drug resistance. The gene expressions of three MCR-related efflux

pumps were assayed; only the mdr1 gene was significantly increased in a

time-dependent manner during culture (Fig 4A). To confirm this result, the protein

expression and functional activities of P-gp were examined, which conclusively

demonstrated high P-gp expression levels in MCF-7 MCLs (Fig 4B-D). These

observations were similar to our previous studies in that ADR induced MCF-7 cell

resistance through the up-regulation of P-gp [34]. However, in our present study,

MCF-7 MCLs were not treated with any drugs, and the increase in P-gp expression

was attributed to the 2D to 3D shift of the culture structure. Pusch et al. [35] also

observed increased P-gp expressions in 3D cultures of intestinal Caco-2 cells.

However, the specific mechanisms were not elucidated.

In our present study, G2/M arrest was observed during the growth of the MCF-7

MCLs (Fig 2D). Emerging evidence has suggested that G2/M arrest could lead to

cancer cell resistance [36-38]. Therefore, we attempted to link G2/M arrest to the

increase of P-gp in MCF-7 MCLs. The expression of Chk2, a controller of G2 arrest,

was found to have significantly increased over time in the MCLs (Fig 5A), especially

on the 14th day (Fig 5B). Chk2 has been reported to phosphorylate p53 to stabilize

p53 [39]. p53 has been described as indispensable for enhanced NF-κB transcriptional

activity [40]. The role of NF-κB in regulating P-gp expression in MCF-7 cells has

been demonstrated in our previous studies [34]. In consideration of these reports, p53

and NF-κB p65 expressions were detected for the MCF-7 MCLs. The results revealed

that the MCF-7 MCL cultures elevated p53 expression and increased p65 expression

in the nuclei (Fig 5A). Accordingly, we hypothesized that G2/M arrest caused the

Page 20 of 45

Accep

ted

Man

uscr

ipt

20

activation of Chk2, which subsequently, increased p53 and promoted p65

accumulation within the nuclei, and finally up-regulated P-gp expression. To confirm

our hypothesis, a Chk2 inhibitor AZD7762 was applied to MCF-7 MCLs to interfere

with G2/M arrest. As anticipated, Chk2, p-Chk2, p53, and p65 expressions

significantly decreased (Fig 5C). More importantly, the initially high expression of

P-gp was also markedly down-regulated (Fig 5D and 5E). Furthermore, the p53

activator Nutlin-3 was found to reverse the down-regulation of p53 and p65 caused by

AZD7762 (Fig 5F), which indicated this pathway was p53-dependent. These data

suggested that P-gp expression during the growth of MCF-7 MCLs was up-regulated

through the Chk2/p53/NF-κB pathway.

In this study, MCF-7 MCLs were used as in vitro models to analyze the

penetration kinetics of ADR to mimic tumor masses in vivo. Due to the different P-gp

expressions when MCF-7 MCLs were cultured for different days, MCF-7 monolayers

and MCLs of 10 and 14 days were chosen as low, medium and high P-gp expression

models to compare the penetration kinetics of ADR and elucidate the influence of

P-gp on drug penetration.

On the one hand, we investigated the different penetration kinetics of ADR in

different cell models. The penetration kinetics of ADR in MCF-7 MCLs of 10 days

were similar to those in MCF-7 monolayers, but were significantly lower than those in

MCF-7 MCLs of 14 days (Fig 6A-C). It was assumed that the penetration kinetics

was determined by two factors: the blocking effect due to the thickness of the

multilayer, and a delivery effect from the P-gp efflux. During the growth of MCLs,

the blocking and delivery effects of P-gp were enhanced. When the MCLs were

cultured for 10 days, these two effects were equivalent, i.e., the “net effect” was zero,

and thus, the penetration kinetics were similar to those in monolayers. When MCLs

Page 21 of 45

Accep

ted

Man

uscr

ipt

21

were cultured for 14 days, the delivery effect from the P-gp was greater than the

blocking effect, and the “net effect” was a delivery effect. Other reasons, such as lack

of oxygen and glucose, may also contribute to the “net effect” [16].

On the other hand, the P-gp inhibitor did not affect the penetration kinetics or

the accumulation of ADR in the MCF-7 monolayer due to the low basal P-gp

expression levels (Fig 6A). However, in the MCF-7 MCLs, the P-gp inhibitor

decreased drug penetration across the MCLs, increased the drug retention within the

cells (Fig 6C and 6D), and further promoted the distribution of ADR across the MCLs

as indicated in the cryosections of MCLs (Fig 6E). This action of the P-gp inhibitor

could be attributed to the high P-gp expression in the MCLs (Fig 4C). The fluorescent

intensity of ADR declined at a 20 μm distance from the top of the MCLs. In the

presence of the P-gp inhibitor, the intensity remained until a 30 μm distance from the

top of the MCLs (Fig 6F), which directly increased the AUC of ADR throughout the

MCLs (Fig 6G). It was speculated that the P-gp aided ADR delivery: ADR entered

the cell by diffusion and was then extruded out of cell by P-gp; subsequently, ADR

was retained in the intercellular space and then diffused into the next cell. This

delivery cycle repeated until ADR reached the bottom of the MCLs. An alternative

mechanism suggests that ADR directly travels to the bottom of the MCLs after

extruding out of a cell into the intercellular space (Fig 8). The drug did not have the

opportunity to interact with its intracellular target nuclei; this may be the reason for

the development of MCR in tumor masses [41, 42]. Hence, blockaded P-gp activity

prevented the intercellular delivery of ADR to the bottom of the MCLs. This

promoted intracellular ADR retention which enabled longer ADR treatment of the

tumor cells. This situation was confirmed in MCF-7 xenografted nude mice in vivo. A

significant promotion of ADR distribution from the outer to inner tumor mass after

Page 22 of 45

Accep

ted

Man

uscr

ipt

22

using the P-gp inhibitor was observed. The P-gp inhibitor helped ADR residing within

the cells from the periphery to the inner core of the tumor mass. The inhibitor also

prevented ADR from being effluxed out of the inner cells and extruded back to the

outer core of the tumor mass through the intercellular spaces. Additionally, the

elevated plasma drug concentration might also contribute to this phenomenon (Fig

7A).

In summary, a 3D-no base and embedded MCF-7 MCL model was successfully

developed for the first time, which provided the quantification of drug penetration

kinetics. During the culture, MCF-7 MCLs gradually became resistant to ADR; this

may be attributed to the up-regulated P-gp expression via the Chk2/p53/NF-κB

pathway. The penetration kinetics of ADR through MCF-7 monolayers and MCLs

were evaluated, compared and mathematically modeled. In combination with

xenografts in vivo, the relationship between the P-gp function and the drug

penetration through the tumor mass was elucidated. Our work provided an applicable

in vitro model to quantitatively evaluate the penetration ability of anti-cancer agents,

and elucidate the role of P-gp in drug delivery through multi-cellular layers. This

model will be helpful for screening new drug candidates / preparations in future and

beneficial for exploring new strategies to overcome MCR in clinical therapy.

Page 23 of 45

Accep

ted

Man

uscr

ipt

23

Acknowledgements

The authors sincerely thank the post-graduates in the Key Lab of Drug Metabolism

and Pharmacokinetics (China Pharmaceutical University, Nanjing, China) for their

kind assistance.

This work was supported by the China National Nature Science Foundation

[No. 81202591]; the Jiangsu Province Nature Science Foundation [No. BK2012354,

BK20131308]; the Fundamental Research Funds for the Central Universities [No.

ZJ13163]; the Jiangsu Province Key Lab of Drug Metabolism and Pharmacokinetics

Projects [No. BM2012012]; the China “Creation of New Drugs” Key Technology

Projects (2015ZX09501001); the Scientific Research and Innovation Project of

College Students of Jiangsu Province [No. CXZZ13_0336]; the Program for New

Century Excellent Talents in University (NCET-11-0740); and the Open Research

Fund of State Key Laboratory of Bioelectronics, Southeast University.

Page 24 of 45

Accep

ted

Man

uscr

ipt

24

Conflicts of interest

The authors declare no conflicts of interest.

Page 25 of 45

Accep

ted

Man

uscr

ipt

25

References

[1] Minchinton AI, Tannock IF. Drug penetration in solid tumours. Nature reviews

Cancer. 2006;6:583-92.

[2] Di Paolo A, Bocci G. Drug distribution in tumors: mechanisms, role in drug

resistance, and methods for modification. Current oncology reports.

2007;9:109-14.

[3] Chitcholtan K, Sykes PH, Evans JJ. The resistance of intracellular mediators to

doxorubicin and cisplatin are distinct in 3D and 2D endometrial cancer. Journal

of translational medicine. 2012;10:38.

[4] Godugu C, Patel AR, Desai U, Andey T, Sams A, Singh M. AlgiMatrix based 3D

cell culture system as an in-vitro tumor model for anticancer studies. PloS one.

2013;8:e53708.

[5] Durand RE, Olive PL. Resistance of tumor cells to chemo- and radiotherapy

modulated by the three-dimensional architecture of solid tumors and spheroids.

Methods in cell biology. 2001;64:211-33.

[6] Solyanik GI. Multifactorial nature of tumor drug resistance. Experimental

oncology. 2010;32:181-5.

[7] Patel KJ, Tannock IF. The influence of P-glycoprotein expression and its

inhibitors on the distribution of doxorubicin in breast tumors. BMC cancer.

2009;9:356.

[8] Carduner L, Picot CR, Leroy-Dudal J, Blay L, Kellouche S, Carreiras F. Cell cycle

arrest or survival signaling through alphav integrins, activation of PKC and

ERK1/2 lead to anoikis resistance of ovarian cancer spheroids. Experimental cell

research. 2014;320:329-42.

Page 26 of 45

Accep

ted

Man

uscr

ipt

26

[9] Evans CJ, Phillips RM, Jones PF, Loadman PM, Sleeman BD, Twelves CJ, et al.

A mathematical model of doxorubicin penetration through multicellular layers.

Journal of theoretical biology. 2009;257:598-608.

[10] Patel KJ, Lee C, Tan Q, Tannock IF. Use of the proton pump inhibitor

pantoprazole to modify the distribution and activity of doxorubicin: a potential

strategy to improve the therapy of solid tumors. Clinical cancer research: an

official journal of the American Association for Cancer Research.

2013;19:6766-76.

[11] Pampaloni F, Reynaud EG, Stelzer EH. The third dimension bridges the gap

between cell culture and live tissue. Nature reviews Molecular cell biology.

2007;8:839-45.

[12] Hirschhaeuser F, Menne H, Dittfeld C, West J, Mueller-Klieser W,

Kunz-Schughart LA. Multicellular tumor spheroids: an underestimated tool is

catching up again. Journal of biotechnology. 2010;148:3-15.

[13] Friedrich J, Seidel C, Ebner R, Kunz-Schughart LA. Spheroid-based drug screen:

considerations and practical approach. Nature protocols. 2009;4:309-24.

[14] Li Q, Chen C, Kapadia A, Zhou Q, Harper MK, Schaack J, et al. 3D models of

epithelial-mesenchymal transition in breast cancer metastasis: high-throughput

screening assay development, validation, and pilot screen. Journal of

biomolecular screening. 2011;16:141-54.

[15] Reid BG, Jerjian T, Patel P, Zhou Q, Yoo BH, Kabos P, et al. Live multicellular

tumor spheroid models for high-content imaging and screening in cancer drug

discovery. Current chemical genomics and translational medicine. 2014;8:27-35.

Page 27 of 45

Accep

ted

Man

uscr

ipt

27

[16] Kyle AH, Huxham LA, Chiam AS, Sim DH, Minchinton AI. Direct assessment

of drug penetration into tissue using a novel application of three-dimensional cell

culture. Cancer research. 2004;64:6304-9.

[17] Hicks KO, Pruijn FB, Secomb TW, Hay MP, Hsu R, Brown JM, et al. Use of

three-dimensional tissue cultures to model extravascular transport and predict in

vivo activity of hypoxia-targeted anticancer drugs. Journal of the National Cancer

Institute. 2006;98:1118-28.

[18] Choi MS, Kim SH, Kuh HJ. Penetration of paclitaxel and 5-fluorouracil in

multicellular layers of human colorectal cancer cells. Oncology reports.

2011;25:863-70.

[19] Zhang J, Zhou F, Wu X, Zhang X, Chen Y, Zha BS, et al. Cellular

pharmacokinetic mechanisms of adriamycin resistance and its modulation by

20(S)-ginsenoside Rh2 in MCF-7/Adr cells. British journal of pharmacology.

2012;165:120-34.

[20] Li L, Lu Y. Optimizing a 3D Culture System to Study the Interaction between

Epithelial Breast Cancer and Its Surrounding Fibroblasts. Journal of Cancer.

2011;2:458-66.

[21] D'Argenio D, Schumitzky A, Wang X. ADAPT 5 User’s Guide:

Pharmacokinetic/Pharmacodynamic Systems Analysis Software. Los Angeles,

CA Biomedical Simulations Resource; 2009.

[22] Zhang J, Zhou F, Wu X, Gu Y, Ai H, Zheng Y, et al. 20(S)-ginsenoside Rh2

noncompetitively inhibits P-glycoprotein in vitro and in vivo: a case for

herb-drug interactions. Drug metabolism and disposition: the biological fate of

chemicals. 2010;38:2179-87.

Page 28 of 45

Accep

ted

Man

uscr

ipt

28

[23] Kim HG, Hien TT, Han EH, Hwang YP, Choi JH, Kang KW, et al. Metformin

inhibits P-glycoprotein expression via the NF-kappaB pathway and CRE

transcriptional activity through AMPK activation. British journal of

pharmacology. 2011;162:1096-108.

[24] Kanagasabai R, Krishnamurthy K, Druhan LJ, Ilangovan G. Forced expression of

heat shock protein 27 (Hsp27) reverses P-glycoprotein (ABCB1)-mediated drug

efflux and MDR1 gene expression in Adriamycin-resistant human breast cancer

cells. The Journal of biological chemistry. 2011;286:33289-300.

[25] LaBarbera DV, Reid BG, Yoo BH. The multicellular tumor spheroid model for

high-throughput cancer drug discovery. Expert opinion on drug discovery.

2012;7:819-30.

[26] Breslin S, O'Driscoll L. Three-dimensional cell culture: the missing link in drug

discovery. Drug discovery today. 2013;18:240-9.

[27] Lee JM, Mhawech-Fauceglia P, Lee N, Parsanian LC, Lin YG, Gayther SA, et al.

A three-dimensional microenvironment alters protein expression and

chemosensitivity of epithelial ovarian cancer cells in vitro. Laboratory

investigation; a journal of technical methods and pathology. 2013;93:528-42.

[28] Kim SH, Kuh HJ, Dass CR. The reciprocal interaction: chemotherapy and tumor

microenvironment. Current drug discovery technologies. 2011;8:102-6.

[29] Doublier S, Belisario DC, Polimeni M, Annaratone L, Riganti C, Allia E, et al.

HIF-1 activation induces doxorubicin resistance in MCF7 3-D spheroids via

P-glycoprotein expression: a potential model of the chemo-resistance of invasive

micropapillary carcinoma of the breast. BMC cancer. 2012;12:4.

Page 29 of 45

Accep

ted

Man

uscr

ipt

29

[30] Yip D, Cho CH. A multicellular 3D heterospheroid model of liver tumor and

stromal cells in collagen gel for anti-cancer drug testing. Biochemical and

biophysical research communications. 2013;433:327-32.

[31] Hicks KO, Siim BG, Jaiswal JK, Pruijn FB, Fraser AM, Patel R, et al.

Pharmacokinetic/pharmacodynamic modeling identifies SN30000 and SN29751

as tirapazamine analogues with improved tissue penetration and hypoxic cell

killing in tumors. Clinical cancer research: an official journal of the American

Association for Cancer Research. 2010;16:4946-57.

[32] Mellor HR, Callaghan R. Accumulation and distribution of doxorubicin in

tumour spheroids: the influence of acidity and expression of P-glycoprotein.

Cancer chemotherapy and pharmacology. 2011;68:1179-90.

[33] Oshikata A, Matsushita T, Ueoka R. Enhancement of drug efflux activity via

MDR1 protein by spheroid culture of human hepatic cancer cells. Journal of

bioscience and bioengineering. 2011;111:590-3.

[34] Zhang J, Lu M, Zhou F, Sun H, Hao G, Wu X, et al. Key role of nuclear

factor-kappaB in the cellular pharmacokinetics of adriamycin in MCF-7/Adr cells:

the potential mechanism for synergy with 20(S)-ginsenoside Rh2. Drug

metabolism and disposition: the biological fate of chemicals. 2012;40:1900-8.

[35] Pusch J, Votteler M, Gohler S, Engl J, Hampel M, Walles H, et al. The

physiological performance of a three-dimensional model that mimics the

microenvironment of the small intestine. Biomaterials. 2011;32:7469-78.

[36] Slupianek A, Hoser G, Majsterek I, Bronisz A, Malecki M, Blasiak J, et al.

Fusion tyrosine kinases induce drug resistance by stimulation of

homology-dependent recombination repair, prolongation of G(2)/M phase, and

protection from apoptosis. Molecular and cellular biology. 2002;22:4189-201.

Page 30 of 45

Accep

ted

Man

uscr

ipt

30

[37] Bao S, Wu Q, McLendon RE, Hao Y, Shi Q, Hjelmeland AB, et al. Glioma stem

cells promote radioresistance by preferential activation of the DNA damage

response. Nature. 2006;444:756-60.

[38] Cavelier C, Didier C, Prade N, Mansat-De Mas V, Manenti S, Recher C, et al.

Constitutive activation of the DNA damage signaling pathway in acute myeloid

leukemia with complex karyotype: potential importance for checkpoint targeting

therapy. Cancer research. 2009;69:8652-61.

[39] Hirao A, Kong YY, Matsuoka S, Wakeham A, Ruland J, Yoshida H, et al. DNA

damage-induced activation of p53 by the checkpoint kinase Chk2. Science.

2000;287:1824-7.

[40] Schneider G, Henrich A, Greiner G, Wolf V, Lovas A, Wieczorek M, et al. Cross

talk between stimulated NF-kappaB and the tumor suppressor p53. Oncogene.

2010;29:2795-806.

[41] Durand RE. The influence of microenvironmental factors during cancer therapy.

In Vivo. 1994;8:691-702.

[42] Primeau AJ, Rendon A, Hedley D, Lilge L, Tannock IF. The distribution of the

anticancer drug Doxorubicin in relation to blood vessels in solid tumors. Clinical

cancer research: an official journal of the American Association for Cancer

Research. 2005;11:8782-8.

Page 31 of 45

Accep

ted

Man

uscr

ipt

31

Tables

Table 1. Features of MCF-7 MCL during culture.

** p<0.01, *** p<0.001 compared with 3-day group.

Culture time

(days)

MCL thickness

(μm)

Packing density

(%)

Cell

layers

Ki-67 positive

expression (%)

3 11.67±1.39 34±6.8 3-4 73±3.2

5 16.59±1.75*** 32±5.0 5-6 80±4.5

8 28.98±1.58*** 32±3.0 6-8 69±5.5

10 34.50±0.87*** 35±3.9 6-9 83±6.1

14 40.17±3.61*** 33±3.6 8-12 89±3.5**

Page 32 of 45

Accep

ted

Man

uscr

ipt

32

Table 2. Kinetics parameters of ADR penetration through MCF-7 MCL with or

without LY335979

* p<0.05 between combined treatment with LY335979 group versus combined

treatment with 0.1% DMSO.

Kinetics

Parameters

0.1% DMSO

group

LY335979

group

F (×10-3, /) 1.36±0.31 1.18±0.22

ka (h-1) 0.033±0.008 0.036±0.007

ke-base ( h-1) 0.022±0.005 0.019±0.005

ke-add ( h-1) 0.21±0.03 0.13±0.03*

ke-induct ( h-1) 0.33±0.04 0.24±0.03*

Page 33 of 45

Accep

ted

Man

uscr

ipt

33

Figure legend

Fig 1 Schematic presentation of an integrated mathematical model describing the

penetration kinetics of ADR through MCLs. c1, c2, and c3 are the concentrations of

ADR in top chamber, multi-cellular layer and bottom chamber, respectively, and ka

and ke are the absorption and elimination rate constants.

Fig 2 Morphology and proliferative features of MCF-7 MCL during the culture time.

MCF-7 cells suspended in serum-free medium containing 6% matrigel were seeded

onto the uncoated cell culture inserts, placed into a 24-well plate and cultured for 3, 5,

8, 10, and 14 days. (A) H&E staining of MCF-7 MCL. (B) Ki-67

immunohistochemical staining of MCF-7 MCL (scale bars represent 25 μm). (C) PI

staining was used to analyze the cell cycle distribution. (D) Percentages of cells in

G2/M phase. Data are presented as means ± S.E., * p<0.05, ** p<0.01 compared with

3-day group.

Fig 3 ADR resistance during the growth of MCF-7 MCL. Cell viability was

determined by MTT assay after cells were exposed to various concentrations of ADR

for 72 h, and IC50 values were calculated. Data are expressed as means ± S.E. of three

independent experiments.

Fig 4 Gene, protein and function levels of P-gp during the growth of MCF-7 MCL. (A)

Relative gene expressions of mdr1, bcrp, and mrp2 by qPCR analysis in MCF-7

monolayer and MCL. Gene expressions were normalized to the housekeeping gene

β-actin, and fold increases was calculated as 3D level/2D level for each gene at

designated culture time. (B) P-gp protein levels were analyzed and quantified by flow

Page 34 of 45

Accep

ted

Man

uscr

ipt

34

cytometry using FITC-conjugated anti-P-gp polyclonal antibody. (C) Analysis of

mean fluorescence intensity of P-gp. (D) Effects of LY335979 on ADR accumulation

in MCF-7 monolayer and MCL. Data are presented as means ± S.E., * p<0.05, **

p<0.01, *** p<0.001 versus 2D group. § p<0.05 between 14-day versus 10-day of

MCL culture. # p<0.05 between combined treatment with LY335979 group versus

combined treatment with 0.1% DMSO for the same MCL culture time.

Fig 5 P-gp up-regulation during the growth of MCL through Chk2/p53/NF-κB

pathway. (A) Western blot assay was performed to detect the expression of p-Chk2,

Chk2, p53 and p65 from whole cell, nuclear and cytoplasmic extracts at designated

culture times. (B) Analysis of protein expression on the 14th day of MCL culture. (C)

Western blot assay was performed to detect the related protein expression on the 14th

day of MCL culture treated with 200 nM and 500 nM AZD7762 for 24 h. (D) P-gp

protein levels of MCF-7 MCL (14-day) treated with 200 nM and 500 nM AZD7762

for 24 h were analyzed and quantified by flow cytometry using FITC-conjugated

anti-P-gp polyclonal antibody. (E) Analysis of mean fluorescence intensity of P-gp. (F)

14th day of MCL culture were pretreated with or without 10 μM Nutlin-3 and then

incubated with 500 nM AZD7762 for 24 h. Western blot was performed to assay the

expression of proteins. Data are presented as means ± S.E., * p<0.05, ** p<0.01, ***

p<0.001.

Fig 6 ADR penetration kinetics evaluated on MCF-7 monolayer and MCL. ADR (10

μM) was applied to the top chamber of the cell culture insert, and samples were

collected at the bottom chamber for MCF-7 monolayer (A), MCL cultured for 10 days

(B) and 14 days (C) in the absence or presence of P-gp inhibitor LY335979. (D)

Page 35 of 45

Accep

ted

Man

uscr

ipt

35

Intracellular retention of ADR within the cells constituting the monolayer or MCL on

the cell culture inserts. (E) Fluorescence imaging of the cryosections of the MCL cells

cultured for 14 days (scale bars represent 40 μm). (F) Plot of fluorescent intensities

against the distance from the top to the bottom of the cell culture insert. (G) AUC of

the intensity-distance curves. Predicted concentration–time curves for ADR

penetration evaluated on MCF-7 monolayer (H) and MCLs cultured for 10 days (I)

and 14 days (J) in the absence or presence of P-gp inhibitor LY335979. Observations

are reported as means ± S.E. (n = 3), and the lines are depicted using the parameters

obtained from the original data. Data are presented as means ± S.E., * p<0.05, **

p<0.01, *** p<0.001 between combined treatment with LY335979 group versus

combined treatment with 0.1% DMSO.

Fig 7 ADR penetration in MCF-7 xenografted nude mice. MCF-7 xenografted nude

mice were administered ADR (30 mg·kg-1, i.v.) in the presence or absence of P-gp

inhibitor LY335979 (25 mg·kg-1, i.v.). 3 h later, concentrations of ADR in plasma (A)

and in tumor (B) were determined. For observation and quantification of ADR in

cryosections of the tumor, cryosections were prepared according to a diagram (C):

with the tumor edge set as 0, the tumor was divided into peripheral region (0-200 μm),

intermediate region (200-400 μm) and central region (400-600 μm). Representative

images (scale bars represent 40 μm) for each region in the absence or presence of

P-gp inhibitor LY335979 was recorded (D), the fluorescence intensity against the

distance was plotted (E), and AUC was calculated (F). Data are presented as means ±

S.E., * p<0.05, ** p<0.01 between ADR and LY335979 combined treatment group

versus ADR treatment group.

Page 36 of 45

Accep

ted

Man

uscr

ipt

36

Fig 8 Schematic representation of proposed cell signal transduction pathways for P-gp

up-regulation through 2D to 3D culture and the potential mechanisms for penetration

kinetics changes of ADR when combined treatment with P-gp inhibitor.

Page 37 of 45

Accep

ted

Man

uscr

ipt

*Graphical Abstract (for review)