You are on page 1of 32

Accepted Manuscript

Title: Assessment of sequential combination of


5-Fluorouracil-loaded-chitosan-nanoparticles and
ALA-Photodynamic therapy on HeLa cell line

Author: Marta Benito-Miguel Dolores Blanco Clara Gómez

PII: S1572-1000(15)00046-0
DOI: http://dx.doi.org/doi:10.1016/j.pdpdt.2015.05.001
Reference: PDPDT 650

To appear in: Photodiagnosis and Photodynamic Therapy

Received date: 24-3-2015


Revised date: 29-4-2015
Accepted date: 4-5-2015

Please cite this article as: Benito-Miguel M, Blanco D, Gómez C, Assessment of


sequential combination of 5-Fluorouracil-loaded-chitosan-nanoparticles and ALA-
Photodynamic therapy on HeLa cell line, Photodiagnosis and Photodynamic Therapy
(2015), http://dx.doi.org/10.1016/j.pdpdt.2015.05.001

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.
Full title: Assessment of sequential combination of 5-Fluorouracil-loaded-chitosan-

nanoparticles and ALA-Photodynamic therapy on HeLa cell line.

Running title: Combination of 5-Fluorouracil-loaded-chitosan-nanoparticles and ALA-

Photodynamic therapy.

t
ip
Authors: Marta Benito-Miguela,bPhD, Mª Dolores Blancob PhD, Clara Gómez*cPhD

cr
a Centro Universitario San Rafael-Nebrija, Madrid, Spain

b Departamento de Bioquímica y Biología Molecular III, Facultad de Medicina, UCM,

us
Madrid, Spain.
c Departamento de Sistemas de Baja Dimensionalidad, Superficies y Materia
Condensada, Instituto de Química Física Rocasolano, CSIC, Madrid,
Spain.

(*) Corresponding author: an


M
Dr. C. Gómez
Departamento de Sistemas de Baja Dimensionalidad, Superficies y Materia Condensada
Instituto de Química Física Rocasolano
Consejo Superior de Investigaciones Científicas, CSIC
d

C/Serrano 119, 28006 Madrid


Tel.: +34-91-561-9400 ext.961252
te

Fax.: +34-91-564-2431
c.gomez@iqfr.csic.es
p
ce

Full title: Assessment of sequential combination of 5-Fluorouracil-loaded-chitosan-


Ac

nanoparticles and ALA-Photodynamic therapy on HeLa cell line.

Running title: Combination of 5-Fluorouracil-loaded-chitosan-nanoparticles and ALA-

Photodynamic therapy.

Authors: Marta Benito-Miguela,bPhD, Mª Dolores Blancob PhD, Clara Gómez*cPhD

a Centro Universitario San Rafael-Nebrija, Madrid, Spain

1
Page 1 of 31
b Departamento de Bioquímica y Biología Molecular III, Facultad de Medicina, UCM,
Madrid, Spain.
c Departamento de Sistemas de Baja Dimensionalidad, Superficies y Materia
Condensada, Instituto de Química Física Rocasolano, CSIC, Madrid,
Spain.

t
ip
(*) Corresponding author:
Dr. C. Gómez
Departamento de Sistemas de Baja Dimensionalidad, Superficies y Materia Condensada

cr
Instituto de Química Física Rocasolano
Consejo Superior de Investigaciones Científicas, CSIC
C/Serrano 119, 28006 Madrid

us
Tel.: +34-91-561-9400 ext.961252
Fax.: +34-91-564-2431
c.gomez@iqfr.csic.es

an
M
d
p te
ce
Ac

HIGHLIGHTS

CNPs were synthesized by the ionic crosslinking method via the TPP addition.

Sequential combination of 5Fu-CNPs and ALA-PDT exhibited the best photocytotoxic


efficiency.

Combination treatment (5Fu-CNPs and ALA-PDT) led to the highest ROS generation.

2
Page 2 of 31
Combination treatment (5Fu-CNPs and ALA-PDT) led to the highest caspase-3
activation.

t
ip
cr
us
an
Abstract

Background: Natural polymers are used as components of nanoparticles (NPs) for drug
M
delivery, as they provide targeted, sustained release and biodegradability.

The purpose of this study was to increase the efficacy of the


d

Photodynamic therapy (PDT) by the combination of 5-aminolevulinic


te

acid (ALA) with 5-Fluorouracil-loaded-chitosan-nanoparticles (5-Fu-


p

CNPs).
ce

Methods: Nanoparticles based on chitosan (CNPs) were synthesized by the ionic

crosslinking method via the TPP addition. 5-Fluorouracil (5-Fu), a first-


Ac

line anticancer drug, was loaded into these 5Fu-CNPs, and they were

assayed as controlled delivery formulation. HeLa cells were incubated in

the presence of 5Fu-CNPs for 24h, next ALA was added to the culture

medium and 4 hours later, to complete the PDT, light irradiation took

place. Analysis of cell viability, reactive oxygen species (ROS)

production, observation of the apoptosis by fluorescence microscopy

followed by analysis of caspase-3 activity were carried out.

3
Page 3 of 31
Results: Spherical 5Fu-CNPs with a mean diameter of 324±43nm, were successfully

synthesized and characterized by TEM and DLS. 5-Fu incorporation was

achieved successfully (12.3 g 5Fu/mg CNP) and the maximum 5-Fu

release took place at 2h. The combined administration of 5Fu-CNPs and

t
PDT mediated by ALA (ALA-PDT) led to an improved efficacy of the

ip
antineoplastic treatment by generation of great cytotoxicity inducted

cr
through an increased ROS production. HeLa cells were destroyed by

apoptosis through activation of caspase pathway.

us
Conclusions: This study proves that combination therapy (photodynamic “ALA” +

an
chemical “5-Fu”+ immunoadjuvant ”chitosan”) may be an effective

approach for the treatment of cancer.


M
Keywords: ALA; Photodynamic therapy; 5-Fu; chitosan; cell viability; ROS.

Introduction
d

The use of NPs in biological applications is being widely explored e.g. polymeric NPs,
te

metallic NPs and quantum dots have been found applicable in drug
p

delivery, bioimaging and biosensing [1]. NPs are utilized for sustained
ce

release of drugs. NPs improve drug efficacy and reduce side effects as a

result of the lower doses that are administered.


Ac

Drug delivery systems based on polymeric NPs, which use natural or synthetic polymer

material have received great attention due to their stability, ease of

surface modification, biocompatibility and biodegradable properties. The

natural polymer chitosan (N-deacetylated derivative of chitin), tends to

be internalized and degraded rapidly, thus enabling a moderate release of

the drug. Chitosan shows more advantages, such as its availability in

natural resources, low cost processing, stability and low toxicity. It has

4
Page 4 of 31
nonspecific antiviral and antitumor activities that have been already

described about three decades ago by Suzuki [2]. Strong systemic and

mucosal immune responses could be induced by chitosan [3]. In this

context, immune-stimulating activity such as increasing accumulation

t
and activation of macrophages and polymorphonuclear cells, inducing

ip
cytokines after intravenous administration, has been reported after

cr
intravenous administration of chitosan [4]. Due to their bioadhesive,

biocompatibility, biodegradability and penetration-enhancement

us
properties, chitosan nanoparticles (CNPs) enhance the immune response

by stimulating the uptake by phagocytotic cells and may also stimulate

an
the immune system [3,5-7]. Therefore, the use of CNPs used as
M
immunological adjuvants to induce both humoral and cell-mediated

immunity could be promising.


d

5-Fluorouracil (5-Fu), is a first –line anticancer drug, which efficacy is affected by its
te

low lipophilicity and low bioavailability and its several side effects [8].

In this manner, small-molecule drugs, as 5-Fu, carried by chitosan or its


p

derivatives, result in extended release, improved bioavailability and


ce

reduced side effects [9]. Due to its pivotal role in the treatment of a
Ac

variety of solid tumors, 5-Fu remains as one of the most commonly used

anticancer drugs and it has been included into a wide range of therapeutic

programs, either alone or in combination with other drugs.

Photodynamic therapy (PDT) is a promising therapeutic procedure for the management

of a variety of solid tumors and non-malignant lesions. It is a treatment

methodology dependent upon administration of a photosensitizer (PS)

and tissue penetrating light of a specific wavelength. When the PS is

5
Page 5 of 31
exposed to light, its molecules become excited and by decaying through

triplet state generate singlet oxygen (1O2) and reactive oxygen species

(ROS) toxic to the tissue cells [10]. The response to the treatment induces

intratumor cytotoxic reaction which involves inflammatory innate,

t
adaptive immune reaction culminating in successful eradication of

ip
residual surviving cancer cells [11]. 5-aminolevulinic acid (ALA) is an

cr
endogenous cellular component and is metabolized within the heme

biosynthetic pathway to produce protoporphyrin IX (Pp IX), a potent

us
endogenous PS. Because ALA is a precursor that bypasses the rate-

limiting enzyme of the heme synthesis pathway (ALA synthase),

an
feedback inhibition of the pathway does not occur and PpIX rapidly
M
accumulates in the cells. Dysplastic cells exhibit decreased ferrochelatase

activity and lower ferric ion concentration, which promotes further


d

accumulation of PpIX than in normal cells. As a result, tumor cells


te

become selectively photosensitized to the action of the light [12].

However, ALA appears to be less effective than other PDT agents


p

(photofrin, etc.) in the case of deep seated tumors. To enhance the


ce

clinical efficacy of ALA-PDT, new methods have been proposed, such as

a combination with hyperthermia or ultrasound, chemotherapy [13,14].


Ac

The efficacy of cancer treatment is not only being increased with the

combination therapy and the dose of photosensitizer and cytostatic drug

can also be reduced without compromising the therapeutic response

[14,15].

In this study, CNPs were prepared by the ionic crosslinking method via the TPP

addition, characterized and evaluated as delivery systems of 5-Fu. Next,

6
Page 6 of 31
5Fu-CNPs were incubated on HeLa cells followed by a photodynamic

treatment mediated by ALA (ALA-PDT), to evaluate the cytotoxic and

apoptotic effect of combined treatment.

t
ip
Materials and methods

cr
Preparation of the 5Fu-CNPs

us
5Fu-CNPs were prepared by the ionic crosslinking method. A dissolution 0.5 % (w/v)

of chitosan in aqueous solution of acetic acid (1% v/v), was added

an
dropwise into 100 mL of 1% (w/v) pH 3 sodium polyphosphate (TPP)

solution. The suspension was sonicated during the time of the addition of
M
chitosan solution. The formation of CNPs started spontaneously via the

TPP initiated ionic gelation mechanism. Then, the suspension was stirred
d

slowly and allowed to crosslink for 1 h at room temperature. Two hours


te

later, the suspension was subject to repeated cycles of washing by


p

deionized water and centrifugation (10,000 rpm for 5 min) to remove the
ce

unreacted chitosan and TPP, then lyophilized and stored at 4 °C.

Fifty milligrams of the above synthesized CNPs were suspended in 1 mL of 10mg/mL


Ac

solution of 5-Fu in deionized water being under slowly magnetic stirring

during 24 h. After this time, the suspension was centrifuged (10,000 rpm

for 1 min), next the suspension was washed in deionized water and

centrifuged again (10,000 rpm for 1 min.). Finally, it was lyophilized and

stored.

Characterization of the 5Fu-CNPs

7
Page 7 of 31
Particle size distribution was determined by dynamic light scattering (DLS) by the size

analyzer (Zetasizer Nano ZS, Malvern Instruments Ltd., Worcestershire,

UK) equipped with a 10 mW helium-neon laser working at a wavelength

of 633 nm, 173° scattering angle and temperature of 25 °C.

t
Measurements were carried out with a suspension of CNPs in water

ip
(0.06% w/v), previously sonicated and incorporated into disposable

cr
cuvettes of polystyrene (Sarstedt AG&Co). The size and distribution of

CNPs were measured by putting the cuvette into the sample cell. Three

us
measurements were made per sample. Morphology and size of the CNPs

was also observed by transmission electron microscopy (TEM) by a

an
JEOL JEM-1010 microscope (JEOL, Korea LTD) at 80 Kv, with a
M
resolution of 0.35 nm. CNPs were dispersed in distilled water and

deposited on 200 mesh Formvar/Carbon grids (Copper, Ted Pella, Inc.


d

Canada). Photographs were analyzed by Megaview Imagine Systems


te

Software (Olimpus Soft Imaging Megaview II Software).


p

In Vitro Release of 5Fu-CNPs


ce

Thirty milligrams of 5Fu-CNPs were suspended in PBS (10 mL, 37ºC, pH = 7.4).

Release study was carried out at a constant temperature (37ºC) and orbital
Ac

shaking (80 rpm) for 24 h by means of an orbital shaker incubator

(Ecotron INFORS HT). At different time intervals 150 L of suspension

were extracted and replaced with 150 L of PBS. The amount of 5-Fu

was measured by UV-vis spectrophotometry at the wavelength of

maximum absorption  max = 266 nm. The drug concentration in each

time is calculated from the calibration curve data to the UV absorption of

8
Page 8 of 31
5 known concentrations of 5-Fu in PBS, in the range from 0.01 to 0.005

mg/mL. Experiments were done in triplicate.

Cell culture

Human cervical cancer (HeLa) cells were maintained in DMEM (4.5 g/L Glucose)

t
ip
supplemented with 10% heat inactivated fetal bovine serum, L-Glutamine

cr
(2mM), penicillin (50 U/mL), streptomycin (50 μg/mL) (Invitrogen Life

Technologies, Grand Island, NY, USA) and gentamicin (50 μg/mL)

us
(Lonza, Basel, Switzerland) at 37ºC under a humidified atmosphere of 5%

CO2 in air. Cells were plated in 75-cm2 flasks (Sarstedt Ag and Co.,

an
Barcelona, Spain) and were passaged upon reaching 95% confluency, by

gentle trypsinization (0.05% trypsin/0.53 mM EDTA; Invitrogen Life


M
Technologies).
d

Photodynamic treatment
te

Experiments were done in triplicate and each experimental condition performed on


p

quintuplicate. HeLa cells were seeded in 96-well flat-bottom plates at


ce

10,000 cells/well. After 24 h, the culture medium was removed and in

some wells CNPs and 5Fu-CNPs were added in the amount needed for
Ac

the release of 5-Fu in the two concentrations studied: 50 and 100 µM.

Two hours later, when 5-Fu release from 5Fu-CNPs was completed, other

wells were incubated with 5-Fu in solution (50 and 100 µM). As control

negative, in some other wells culture medium were added. All the wells

were incubated for 24 h, and next, the cells were washed twice with 100

L of serum-free DMEM. Afterward, ALA in solution in serum-free

DMEM (50 and 75 µM) was added and then cells were re-incubated for 4

9
Page 9 of 31
DMEM (50 and 75 µM) was added and then cells were re-incubated for 4

h before irradiation. Cells were irradiated using a LED Therapy Beauty

Equipment (L-35) (Guangzhou JIMY, China) (=640 nm, 40 mW cm-2)

applied at a distance of 8 cm from the cells during 7 min. After irradiation

t
treatment, 10 L of FBS were added to each well.

ip
Cytotoxicity

cr
Cell viability was analyzed by MTT assay, method based on the activity of

us
mitochondrial dehydrogenases. MTT assay was deemed a suitable test for

cell viability determination after PDT photoactivation. MTT (3-(4,5-

an
dimethylthiazol-2-yl)-2,5-diphenyltertrazolium bromide; Sigma-Aldrich)

is reduced to a purple insoluble formazan derivative in living,


M
metabolically active cells.
d

After PDT cells were incubated for 20h before the MTT assay was performed. Fifty
te

microliters of MTT solution (concentration 1 mg/mL) were added to each

well and the plate re-incubated at 37ºC for 2 h. MTT was then replaced
p

with 100 μL DMSO (Sigma-Aldrich). Cell viability was determined by


ce

measuring the absorbance at 570 nm using a microplate reader

(Varioskan, Thermo Fisher Scientific, Barcelona, Spain). Results are


Ac

presented as the percentage of surviving cells in relation to that of

untreated control cells.

ROS production

Using the fluorescent probe 2´,7´-dichlorodihydrofluorescein diacetate (H2DCFDA)

(Molecular Probes/Invitrogen), the endogenous ROS production was

10
Page 10 of 31
quantified. Non-fluorescent H2DCFDA is converted to its fluorescent

2´,7´-dichlorofluorescein (DCF) by ROS species. The fluorescent

compound can be quantified using a Varioskan microplate reader. Cells

were pre-treated with H2DCFDA (Molecular Probes/Invitrogen) (100L,

t
40 M) 10 min before ALA addition. Four hours later, immediately after

ip
PDT, the well plate was incubated for a further 1 h at 37ºC. Next, culture

cr
medium was removed and PBS was added (as culture medium interfered

the measurement). ROS fluorescence intensity (resulting from the

us
oxidation of dye), was then measured (excitation wavelength at 488/12

an
nm and emission wavelength at 520 nm). Then, MTT assay was carried

out to calculate ROS production per life cell.


M
Observation of the apoptosis by fluorescence microscopy

Cells were seeded onto cover glasses, positioned in p24 at a concentration of 50,000
d

cell/well in 1 mL of growth medium and then treated with the same


te

protocol as described for PDT. Twenty minutes after irradiation, cells


p

were washed with PBS and Annexin V and Propidium iodide (PI) were
ce

added according the manufacturer´s instructions (BD Pharmingen,

Annexin V-FITC Apoptosis Detection Kit I; BD Biosciences, San Diego,


Ac

CA, USA). Fluorophores were incubated during 15 min. in dark and at

room temperature. Then the medium was discarded. The cells were fixed

for 10 minutes in 1% paraformaldehyde in PBS, and mounted onto glass

slides with Fluoromount G® (Souther Biotechnology Associates, Inc.).

To observe apoptosis and necrosis, images of stained cells were taken by

using a TSC SP2 confocal laser microscopy system (Leica, Wetzlar,

11
Page 11 of 31
Germany) equipped with an inverted DMIRE2 Leica microscope.

Fluorescence images were analyzed using LCS software from Leica.

Caspase-3 activity assay

HeLa cells were growth in p24 at a concentration of 100,000 cell/well in 1 mL of growth

t
ip
medium. Briefly, 24 h after PDT treatment, each cell conditions were

cr
collected in 150 L of lysis buffer and centrifuged at 13,000 rpm, 4ºC

during 15 min. Then the supernatant was stored at -80ºC. Estimation of

us
total protein in the supernatant was performed by Bradford method before

running the caspase-3-activity assay with 40 g of the protein extract. The

fluorogenic caspase-3

an
substrate (Caspase 3 Ac-DEVD-AMC,

Calbiochem; Alexis- Enzo Life Sciences) was added and incubated during
M
1 h at 37ºC. Using the Varioskan microplaque reader, levels of cleaved

caspase substrate were monitorized at excitation/emission = 380 nm/460


d

nm. Caspase-3 activity was normalized to total protein content and


te

expressed as fluorescence intensity in arbitrary units per mg protein.


p

Statistical analysis
ce

All data were expressed as the mean± SD of three independent experiments. Statistical
Ac

analyses and calculations were performed with SPSS for Windows

software (version 15.0; SPSS, Chicago, III). Differences between groups

were analyzed using the analysis of variance (ANOVA). Previously, the

normality of the data was checked by using the Shapiro-Wilk test. Values

of p<0.05 and p<0.001 were considered statistically significant and very

statistically significant, respectively.

12
Page 12 of 31
Results

Characterization of CNPs and 5-Fu release from CNPs

t
TEM micrographs of 5Fu-CNPs showed individual and independent particles of very

ip
small size, most of them around ~ 18-20 nm with a very homogeneous

cr
morphology and spherical shape (Figure 1A), but usually CNPs tend to

form aggregates up to almost 200 nm (Figure 1B). DLS technique was

us
also applied to investigate the average size of the particles, resulting in an

an
average diameter of 324±43nm (Figure 1C). It should be pointed out that

the particle size determined by DLS technique is a hydrodynamic


M
diameter. The larger diameter showed by DLS technique in contrast to

TEM measures might be a result of the aggregation of single CNPs while


d

dispersed in water.
te

Drug release studies were carried out at 37ºC. 5-Fu release from CNPs is shown in
p

Figure 1D. Maximum 5-Fu release took place 2 hour after starting the
ce

experiment. The amount of drug incorporated was: 12.3 g 5-Fu/mg

CNP.
Ac

Cell viability

To explore whether CNPs, 5Fu-CNPs, 5-Fu and ALA-PDT have positive interactions

with mortality of Hela cells when given in combination, it is necessary to

determine their own cytotoxicity. The 5-Fu doses examined were 50 and

100 M and 50 and 75 M for ALA. In the dark, all conditions without 5-

Fu demonstrated a consistent and low cytotoxicity (Figure 2A). 5-Fu

13
Page 13 of 31
exerts its cytotoxic action in a way similar both in solution and loaded

into the CNPs and higher at the highest dose (100M).

After 7 min of PDT, all conditions with ALA were affected to a greater extent with the

highest dose of ALA applied (75M). The combination treatment, either

t
ip
based on 5-Fu loaded into CNPs with ALA-PDT or 5-Fu solution with

ALA-PDT, enhances the final antitumorigenic effect, lowering decreasing

cr
doses of both 5-Fu and ALA and thus minimizing collateral effects

us
(Figure 2B). Combination treatment significantly enhances the percentage

of cell death in comparison with the individual treatment even with the

an
use of the lower doses (50 M for 5-Fu; 50 M for ALA) thereby

enabling reducing the doses and thus side effects. In addition, a slightly
M
more effective effect was observed when 5-Fu was administered loaded

into CNPs than in solution into the combined treatment. To obtain this
d

therapeutic reinforcement chemotherapy mediated by 5-Fu should be


te

applied 24 h before ALA application and not concomitantly (see


p

Supplementary Fig. 1, Supplemental Digital Content 1).


ce

Figure 2C shows how chitosan enhances (but not significantly) the effect of ALA. PDT

triggers an inflammatory response and enhances a specific antitumor


Ac

response that could be further stimulated by the addition of adjuvants as

chitosan.

Reactive oxygen species levels

Figure 3 shows how ROS production can be correlated with the results shown in Figure

2B. At greater decrease in cell viability higher increased production of

14
Page 14 of 31
ROS was observed. Pretreatment with 5-Fu, augmented the expected

response of ROS production (which was higher when using 5Fu-CNPs

than with 5-Fu solution), due to ALA-PDT treatment. It is important to

point out that the individual treatment with 5-Fu either in solution or

t
incorporated into CNPs was able to generate ROS.

ip
Observation of the apoptosis by fluorescence microscopy

cr
To prove that combination treatment reduced cell viability by inducing apoptosis, we

us
investigated apoptotic cells through fluorescence microscopy by applying

Annexin V and Propidium iodine (AV/PI) double staining method.

an
Results after light irradiation are shown in Figure 4, where late apoptotic

cells (AV+/PI+) were visualized, in increasing order of the number of


M
apoptotic cells from CNPs (top), followed by the individual treatments

with 5-Fu, 5Fu-CNPs and ALA (middle), to the combination treatment


d

(5-Fu solution plus ALA and 5-Fu loaded into CNPs plus ALA) (bottom).
te

Caspase-3 activity assay


p
ce

Apoptosis can also be analyzed from a quantification of the enzymatic activity of

caspases. Therefore, the activity of the caspase-3 as apoptotic marker was


Ac

determined (Figure 5). Results showed that the activity of caspase-3 was

higher after the combination treatment (although very similar for both

types of combination treatment: solution of 5-Fu with ALA-PDT and

5Fu-CNPs with ALA-PDT), than with the individual treatments applied.

15
Page 15 of 31
Discussion

In the present study, CNPs were used to deliver 5-Fu. The ionic gelation method was

followed to prepare CNPs using medium molecular weight chitosan. The

size of the CNPs ranged from 281 to 367 nm. Previous studies revealed

t
ip
that CNPs could be used for delivery of various biological molecules and

drugs [16,17]. 5-Fu is a widely used anticancer drug that is toxic to

cr
normal cells and has a short plasma half-life of 15-20 min. In the present

us
study, 5Fu-CNPs were synthesized with the aim of prolonging drug action

and thus improving therapeutic results. 5-Fu loaded into NPs based in

an
chitosan could improve the bioavailability, the site-specific of delivery

and reduction of its side-effects providing obvious therapeutic advantages


M
[9,18].

The effects of ALA-PDT and possible mechanisms involved in the treatment of cervical
d

cancer in vitro and in vivo have been already explored [19,20]. PDT has
te

several features that distinguish it among other antitumor therapies. Its


p

unique mechanism of action include direct cytotoxic effects exerted


ce

towards tumor cells (lipid peroxidation, crosslinking of proteins and

damage to multiple cellular sites, including: membranes, DNA, and


Ac

cytoskeleton), destruction of tumor and peritumoral vasculature and

induction of inflammatory innate, adaptive immune reaction culminating

in successful eradication of residual surviving cancer cells [21,22]. For

clinical applications, an ideal PDT should exhibit extensive killing of the

tumor cells with minimal damage to host tissues in the area of tumor. It is

still possible to damage neighboring healthy cells if the PS dose is too

16
Page 16 of 31
high. It would be ideal if a lower PS dose that is safe for healthy human

tissues can efficiently function as a tumoricide agent.

The potential benefits of combination therapies including PDT in association with other

pharmacological therapies over monotherapy for the treatment of several

t
ip
types of cancer, has been supported by some studies [13,15,23] PDT in

combination with chemotherapeutic agents has shown an increase of the

cr
direct damage to the targeted cells and thus an increase of the cytotoxicity

us
by modulating signal pathways that may lead to apoptosis, alterations of

the tumor microvasculature and induction of a host immune response

an
against cancer cells [23].

At least three different ways in which the 5-Fu can exert antitumor action have been
M
described: inhibition of thymidylate synthase (enzyme essential for the

synthesis of the majority of thymidine nucleotides, precursor necessary in


d

DNA synthesis); inhibition of RNA function and protein synthesis; and


te

incorporation into DNA leading to their break [24,25]. Some studies also
p

have shown that pro-oxidant actions could enhance the antitumor efficacy
ce

of 5-Fu [26,27]. Therefore, it is expected that 5-Fu increases its

effectiveness when it is combined with therapies that induce oxidative


Ac

effect as PDT.

Chitosan was applied to further enhance the immune response induced by PDT. By

stimulating inflammation and generating apoptotic and necrotic tissue,

PDT provides an ideal environment for initiation of an antitumor immune

response. Addition of adjuvants leads to PDT enhancement of the

17
Page 17 of 31
photodynamic effect [28,29].

In this work the efficacy of a combined treatment based on 5Fu-CNPs and ALA-PDT

was studied. HeLa cells were utilized as in vitro model of human cervical

cancer. From the MTT assay, 5-Fu had the same cytotoxic effect with and

t
ip
without light irradiation and higher at the highest dose. CNPs were not

cytotoxic with and without light irradiation, but enhanced the

cr
photodynamic effect of ALA. Combination of ALA-PDT with 5-Fu

us
enhanced the percentage of cell death, and a slightly more when 5-Fu was

released from CNPs than by using 5-Fu in solution. Therefore, the

an
administration of CNPs shows a slight added value. PDT leads to direct

tumor cell destruction and serves as the precursor for host immune
M
responses by exposing tumor antigens [28]. Chitosan in combination with

PDT could stimulate and enhance the host immune system to mount a
d

direct attack, with the help of the exposed antigens, against the remaining
te

tumor cells [29]. CNPs could be outlined to be an interesting complement

of PDT.
p
ce

To maximize the effect of the combination treatment, chemotherapy mediated by 5-Fu

should be applied a day before ALA application (to ensure that the 5-Fu
Ac

has exerted its action). However, simultaneous application of 5-Fu and

ALA did not result in a more effective therapy (see Supplementary Fig. 1,

Supplemental Digital Content 1).

ROS have been shown to destroy tumors by three distinct mechanisms: (1) direct

cytotoxicity towards tumor cells producing apoptosis and necrosis; (2)

destruction of tumor capillaries and microvasculature mediated by PDT

18
Page 18 of 31
damage towards endothelial cells; (3) an acute inflammatory response that

can activate host-defense mechanisms involving neutrophils and dendritic

cells [30]. In addition, it has been demonstrated that ROS generation is

higher in cells treated with PDT compared to the normal untreated cells

t
[31]. As PDT belongs to treatment modalities that generate ROS

ip
overproduction, generation of ROS was also analyzed in the present

cr
study. In addition, 5-Fu generates oxidative stress while its action is

favored in a pro-oxidant cellular environment as generated by PDT

us
[26,27]. In our study, ROS production was higher in the cells treated with

ALA-PDT, (and higher at the highest concentration of ALA tested, 75

an
M) compared to the treatment without ALA application. The generation
M
of ROS in the HeLa cells treated with the combined treatment was higher

than the observed after the individual treatments (ALA, CNPs, 5-Fu in
d

solution, 5Fu-CNPs). This effect was more evident in the 5Fu-CNPs and
te

ALA-PDT combined treatment.


p

Cells undergoing apoptosis display typical features. Dramatic nuclear changes are
ce

common during apoptotic death due to activation of endogenous

nuclease(s) which cleaves DNA into oligonucleosomal fragments.


Ac

Chromatin condensation, nuclear shrinkage and formation of apoptotic

bodies can easily be observed under fluorescence microscopy, after

appropriate staining of nuclei with DNA-specific fluorochromes. Annexin

and PI double staining were performed to determine if the combination

treatment had an enhanced apoptotic effect compared to the individual

treatments. It has been reported that PDT with mitochondrial localizing

PS, such as ALA, can induce rapid cell death apoptosis [32,33].

19
Page 19 of 31
PS, such as ALA, can induce rapid cell death via apoptosis [32,33].

Results of the present work showed that the number of late apoptotic cells

visualized by fluorescence microscopy increased in cells treated with

individual treatments and even more in cells treated with combination

t
treatment. These results were in agreement with those obtained from the

ip
cell viability analysis and ROS quantification.

cr
Insult or failure of the mitochondria could rapidly lead to the inhibition of cell survival

us
and proliferation, therefore the induction of mitochondrial impairment

may be a successful anticancer strategy. The opening of the mitochondrial

an
permeability transition pore can lead to a release of pro-apoptotic

molecules from the intermembrane space causing the activation of


M
caspase-9, which further activates caspase-3 [34]. Caspase-3, is a member

of the caspase family of 13 aspartate specific cysteine proteases that plays


d

a central role in the execution of the apoptotic program [35]. To find out
te

the possible pathway of the enhanced apoptosis observed after the

combination treatment, the expression level of caspase-3 was studied.


p

Results showed that enzyme activity of caspase-3 was higher in case of


ce

combination treatment compared to the control cells as well as the


Ac

individual treatments. Increased caspase-3 activity leads to apoptosis and

finally cancer cell destruction. ROS are significant regulators of apoptosis

and activation of apoptosis is associated with generation of ROS. In this

study, combined treatment destroyed HeLa cells through an increased

ROS generation which mediates apoptosis by enhancing the activity of

caspase-3.

20
Page 20 of 31
Conclusion

With this combination treatment, not only a higher efficacy can be achieved but also

both the effective dose of the chemotherapeutic drug and photosensitizer

t
ip
can be reduced, and hence, the adverse side effects of the drugs used can

cr
also be controlled. Immunoadjuvant contribution cooperates positively in

the final result. Further studies for assessing the efficacy and risk-benefit

us
of that PDT-combined treatment are needed to establish this combination

treatment as alternative for malignant disorders.

an
M
Acknowledgements/ Conflicts of Interest and Source of Funding: This research was
d

supported by a Grant from “UCM Group-Santander Bank (Group


te

920613)” and by the Spanish Research Project MICINN (MAT2010-


p

20646-C04-01). The authors have no other relevant affiliation or financial


ce

involvement with any organization or entity with a financial interest in or

financial conflict with the subject matter or materials discussed in the


Ac

manuscript apart from those disclosed.

References

[1] Fröhlich E. The role of surface charge in cellular uptake and cytotoxicity of medical
nanoparticles. Int J Nanomed 2012; 7: 5577-91.

[2] Suzuki, K, Okawa Y, Hashimoto K, Suzuki S, Suzuki M. Protecting effect of chitin


and chitosan on experimentally induced murine candidiasis. Microbiol
Immunol 1984; 28: 903-12.

[3] Zhu B, Qie Y, Wang J, Zhang Y, Wang Q, Xu Y, et al. Chitosan microspheres

21
Page 21 of 31
enhance the immunogenicity of an Ag85B-based fusion protein
containing multiple T-cell epitopes of Mycobacterium tuberculosis. Eur J
Pharm Biopharm 2007; 66: 318–26.

[4] Seferian PG, Martínez ML. Immune stimulating activity of two new chitosan
containing adjuvant formulations. Vaccine 2000; 19: 661–8.

[5] Zhao Y, Park RD, Muzzarelli RA. Chitin deacetylases: properties and applications.

t
Mar Drugs 2010; 8: 24-46.

ip
[6] Arias JL, Reddy LH, Couvreur P. Superior preclinical efficacy of gemcitabine

cr
developed as chitosan nanoparticulate system. Biomacromolecules 2011;
12: 97-104.

us
[7] Joshi VB, Geary SM, Salem AK. Biodegradable particles as vaccine antigen delivery
systems for stimulating cellular immune responses. Hum Vaccin
Immunother 2013; 9: 2584-90.

an
[8] Ofverholm A, Arkblad E, Skrtic S, Albertsson P, Shubbar E, Enerbäck C. Two cases
of 5-fluorouracil toxicity linked with gene variants in the DPYD gene.
Clin Biochem 2010; 43: 331-4.
M
[9] Cheng M, Gao X, Wang Y, Chen H, He B, Xu H, et al. Synthesis of glycyrrhetinic
acid-modified chitosan 5-fluorouracil nanoparticles and its inhibition of
liver cancer characteristics in vitro and in vivo. Mar Drugs 2013; 11:
d

3517-36.
te

[10] Palumbo G. Photodynamic therapy and cancer: a brief sightseeing tour. Expert Opin
Drug Deliv 2007; 4: 131-48.
p

[11] Brown SB, Brown EA, Walker I. The present and future role of photodynamic
therapy in cancer treatment. Lancet Oncol 2004; 5: 497-508.
ce

[12] Castano AP, Demidova TN, Hamblin MR. Mechanisms in photodynamic therapy:
part one-photosensitizers, photochemistry and cellular localization.
Photodiagn Photodyn Ther 2004; 1: 279-93.
Ac

[13] Nonaka M, Ikeda H, Inokuchi T. Effect of combined photodynamic and


chemotherapeutic treatment on lymphoma cells in vitro. Cancer Lett
2002; 184: 171-8.

[14] Zuluaga MF, Lange N. Combination of photodynamic therapy with anti-cancer


agents. Curr Med Chem 2008; 15: 1655-73.

[15] Ahn JC, Biswas R, Chung PS. Combination with genistein enhances the efficacy of
photodynamic therapy against human anaplastic thyroid cancer cells.
Laser Surg Med 2012; 44: 840-9.

22
Page 22 of 31
[16] Rojanarata T, Petchsangsai M, Opanasopit P, Ngawhirunpat T, Ruktanonchai U,
Sajomsang W, Tantayanon S. Methylated N-(4-N,N-
dimethylaminobenzyl) chitosan for novel effective gene carriers. Eur J
Pharm Biopharm 2008; 70: 207–14.

[17] Lam TD, Hoang VD, Lien LN, Thinh NN, Dien PG. Synthesis and characterization
of chitosan nanoparticles used as drug. Vietnam J Chem 2006; 44: 105–9.

t
[18] Li G, Guo L, Wen Q, Zhang T. Thermo-and pH-sensitive ionic-crosslinked hollow

ip
spheres from chitosan-based graft copolymer for 5-fluorouracil release.
Int J Biol Macromol 2013; 55: 69-74.

cr
[19] He GF, Bian ML, Zhao YW, Xiang Q, Li HY, Xiao C. Apoptosis-inducing effect of
5-aminolevulinic acid-mediated photodynamic therapy (5-ALA-PDT) on

us
cervical cancer cell lines. Ai Zheng 2008; 27: 897-904.

[20] He GF, Bian ML, Zhao YW, Xiang Q, Li HY, Xiao C. A study on the mechanism
of 5-aminolevulinic acid photodynamic therapy in vitro and in vivo in

an
cervical cancer, Oncol Rep 2009; 21: 861-8.

[21] Anand S, Ortel BJ, Pereira SP, Hasan T, Maytin EV. Biomodulatory approaches to
M
photodynamic therapy for solid tumors. Cancer Lett. 2012; 326: 8-16.

[22] Milla Sanabria L, Rodríguez ME, Cogno IS, Rumie Vittar NB, Pansa MF,
Lamberti MJ, Rivarola VA. Direct and indirect photodynamic therapy
d

effects on the cellular and molecular components of the tumor


microenvironment. Biochim Biophys Acta 2013; 1835: 36-45.
te

[23] Nonaka Y, Nanashima A, Nonaka T, Uehara M, Isomoto H, Abo T, Nagayasu T.


Synergic effect of photodynamic therapy using talaporfin sodium with
p

conventional anticancer chemotherapy for the treatment of bile duct


carcinoma. J Surg Res 2013;181: 234-41.
ce

[24] Chu E, Mota AC, Fogarasi MC. Antimetabolites, In: DeVita VT, Hellman S,
Rosenberg SA, eds. Cancer: Principles and Practice of Oncology, 6th
Ac

edition, Philadelphia: Lippincott Williams &Wilkins, 2000: 394-8.

[25] Hernández-Vargas H, Ballestar E, Carmona-Saez P, von Kobbe C, Bañón-


Rodríguez I, Esteller M, Moreno-Bueno G, Palacios J. Transcriptional
profiling of MCF7 breast cancer cells in response to 5-Fluorouracil:
Relationship with cell cycle changes and apoptosis, and identification of
novel targets of p53. Int J Cancer 2006; 119: 1164-75.

[26] Chibber S, Farhan M, Hassan I, Naseem I. White light-mediated Cu(II)-5Fu


interaction augments the chemotherapeutic potential of 5-Fu: an in vitro
study. Tumour Biol 2011; 32: 881-92.

[27] Fu Y, Yang G, Zhu F, Peng C, Li W, Li H, et al. Antioxidants decrease the

23
Page 23 of 31
apoptotic effect of 5-Fu in colon cancer by regulating Src-dependent
caspase-7 phosphorylation. Cell Death Dis 2014; 5, e983;
doi:10.1038/cddis.2013.509.

[28] St. Denis TG, Aziz K, Waheed AA, Huang Y-Y, Sharma SK, Mroz P, Hamblin
MR. Combination approaches to potentiate immune response after
photodynamic therapy for cancer. Photochem Photobiol Sci 2011; 10:
792-801.

t
ip
[29] Chen WR, Korbelik M, Bartels KE, Liu H, Sun J, Nordquist RE. Enhancement of
laser cancer treatment by a chitosan-derived immunoadjuvant. Photochem
Photobiol 2005; 81: 190‒5.

cr
[30] Castano AP, Mroz P, Hamblin MR. Photodynamic therapy and anti-tumour

us
immunity. Nat Rev Cancer 2006; 6: 535-45.

[31] Sharma S, Jajoo A, Dube A. 5-aminolevulinic acid-induced protoporphyrin-IX


accumulation and associated phototoxicity in macrophages and oral

an
cancer cell lines. J Photochem Photobiol B 2007; 88: 156-62.

[32] Tsai JC, Wu CL, Chien HF, Chen CT. Reorganization of cytoskeleton induced by
5-aminolevulinic acid-mediated photodynamic therapy and its correlation
M
with mitochondrial dysfunction. Lasers Surg Med 2005; 36: 398-408.

[33] Tsai T, Ji HT, Chiang PC, Chou RH, Chang WS, Chen CT. ALA-PDT results in
phenotypic changes and decreased cellular invasion in surviving cancer
d

cells. Lasers Surg Med 2009; 41: 305-15.


te

[34] Launay S, Hermine O, Fontenay M, Kroemer G, Solary E, Garrido C. Vital


functions for lethal caspases. Oncogene 2005; 24: 5137-48.
p

[35] Cohen G. Caspases: the executioners of apoptosis. Biochem J 1997; 326(Pt 1): 1-
ce

16.
Ac

FIGURE LEGENDS

24
Page 24 of 31
Figure 1.- (A,B) TEM images of the CNPs. (C) Particle size distribution histogram of

CNPs obtained by DLS data. (D) Cumulative amount of 5-Fu released

from 5Fu-CNPs at 37ºC. Each value represents the mean±SD (n=3).

Figure 2.- (A) Viability of HeLa cells in percentage determined by the MTT assay in

t
ip
dark conditions. Each value represents the mean±SD of the three

independent experiments. (*) Statistically significant differences with a

cr
significant decrease from values of control cells (p<0.05). Combined

us
treatment was performed at low doses (5-Fu 50 M and ALA 50 M) and

at high doses (5-Fu 100 M and ALA 75 M). (B) Viability of HeLa

an
cells after PDT treatment in which cells were irradiated for 7 min under

40mWcm-2 led lamp. Each value represents the mean±SD of the three
M
independent experiments. (*) p<0.05 and (**) p<0.001 Statistically and

very statistically significant differences, respectively, with a significant


d

decrease from the same treatment at dark conditions. (##) p<0.001 Very
te

statistically significant differences with a significant decrease from ALA


p

group. Combined treatment was performed at low doses (5-Fu 50 M


ce

and ALA 50 M) and at high doses (5-Fu 100 M and ALA 75 M).

Figure 2C shows the effect of chitosan as adjunctive agent in the PDT


Ac

mediated by ALA. Each value represents the mean±SD of the three

independent experiments. (*) p<0.05 and (**) p<0.001 Statistically and

very statistically significant differences, respectively, with a significant


ns
decrease from values of control cells. ( ) Not significant differences

between ALA+light and CNPs+ALA+light groups.

Figure 3.- ROS production after PDT treatment. Values have been normalized by values

25
Page 25 of 31
of living cells. Data represent the mean±SD of the three independent

experiments. (*) p<0.05 and (**) p<0.001 Statistically and very

statistically significant differences, respectively, with a significant

increase from values of control cells.

t
ip
Figure 4.- Fluorescence microscope images of HeLa cells incubated with Annexin V

and PI 20 min after PDT for visualization by fluorescence microscopy.

cr
Experiment was done with the highest concentrations of ALA and 5-Fu

us
used in the present study (75 M and 100 M, respectively). The green

fluorescence on the cells indicated apoptotic cells, the red fluorescence on

an
the cells indicated late apoptotic cells. Scale bar: 100µm.

Figure 5.- Activity of caspase 3 in cell lysates. Experiment was done with the highest
M
concentrations of ALA and 5-Fu used in the present study (75 M and

100 M, respectively). Data represent the mean±SD of the three


d

independent experiments. (*) p<0.05 and (**) p<0.001 Statistically and


te

very statistically significant differences, respectively, with a significant


p

increase from the same treatment at dark conditions.


ce
Ac

SUPPLEMENTAL DIGITAL CONTENT

Supplemental Digital Content 1.pdf

26
Page 26 of 31
Figure

t
ip
cr
us
an
M
d
te
p
ce
Ac

Page 27 of 31
Figure

t
ip
cr
us
an
M
d
te
p
ce
Ac

Page 28 of 31
Figure

i
cr
us
an
M
ed
pt
ce
Ac

Page 29 of 31
Figure

t
ip
cr
us
an
M
d
te
p
ce
Ac

Page 30 of 31
Figure

i
cr
us
an
M
ed
pt
ce
Ac

Page 31 of 31

You might also like