The International Journal of Biochemistry & Cell Biology

You might also like

Download as pdf or txt
Download as pdf or txt
You are on page 1of 14

The International Journal of Biochemistry & Cell Biology 40 (2008) 2192–2205

Contents lists available at ScienceDirect

The International Journal of Biochemistry


& Cell Biology
journal homepage: www.elsevier.com/locate/biocel

Cellular inactivation and antitumor efficacy of a new zinc


phthalocyanine with potential use in
photodynamic therapy
Natalia B. Rumie Vittar a,1 , Cesar G. Prucca a,1 , Cristian Strassert b ,
Josefina Awruch b , Viviana A. Rivarola a,∗
a
Departamento de Biologı́a Molecular, Facultad de Ciencias Exactas, Fı́sico-Quı́micas y Naturales,
Universidad Nacional de Rı́o Cuarto, Agencia Postal Nro 3, 5800 Rı́o Cuarto, Córdoba, Argentina
b
Departamento de Quı́mica Orgánica, Facultad de Farmacia y Bioquimica, Universidad de Buenos Aires,
Junı́n 956, 1113 Buenos Aires, Argentina

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

Article history: The aim of the present study was to evaluate the photodynamic efficacy of a
Received 26 August 2007 novel phthalocyanine derivate 2,3,9,10,16,17,23,24-octakis[(N,N-dimethylamino) ethylsul-
Received in revised form 18 February 2008
fanyl]phthalocyaninatozinc(II) (referred here as S1) using MCF-7c3 human breast cancer
Accepted 27 February 2008
cells and the LM2 adenocarcinoma subcutaneously implanted in Balb/c mice as experi-
Available online 6 March 2008
mental models.
The S1-l-␣-dimyristoyl-phosphatidylcholine liposome was selected as the best delivery
Keywords:
Photodynamic Therapy system because it showed greater internalization into cells (35 nmol/106 cells), relative to
Photosensitizer other liposomes. After 3 h incubation S1 was partially localized in lysosomes, the compart-
Apoptosis ment that represented its primary photodamage site. The S1 treated cultures also revealed
Necrosis a degree of mitochondrial morphology alteration. Indeed, S1 leads to photokilling of the
Cancer cells with different efficacies indicating that cell photoinactivation was dependent on both
the phthalocyanine concentration and the light dose applied. Analyses of morphology and
nuclear condensation level indicated that some of the cells exposed to photodynamic ther-
apy were undergoing apoptosis within 8 h after treatment.
To assess the in vivo effectiveness of S1, animals bearing tumors were treated with
0.2 mg/kg S1 followed 24 h later by 108 J cm−2 light at 600–800 nm and 60 mW cm−2 ,while
other animals served as controls (no treatment, light alone, or S1 alone). All S1 treated
tumors and none of the controls exhibited complete or partial responses, and these
responses continued for the entire observation period of 12 days. Evaluation of tumor size
showed that the treatment effectively delayed tumor growth. Light microscopy investi-
gations of irradiated tumor specimens showed that S1 causes an early direct damage of
malignant cells, largely via processes leading to random necrotic pathways.
© 2008 Elsevier Ltd. All rights reserved.

Abbreviations: PDT, photodynamic therapy; Pc, phthalocyanine; PS, photosensitizer; DBU, 1,8-diazabicyclo[5.4.0]undec-7-ene; DMPC, l-␣-
dimyristoyl-phosphatidylcholine; DPPEc, d,l-␣-dipalmitoyl phosphatidylethanolamine containing cholesterol; PBS, phosphate-buffered saline; SDS,
sodium dodecyl sulfate; DMF, dimethylformamide; THF, tetrahydrofuran; HO258, Hoechst33258; MTT, 3-(4,5-dimethylthiazolil-2)-2,5
diphenyltetrazolium bromide; H&E, hematoxylin-eosin.
∗ Corresponding author. Tel.: +54 358 4676437; fax: +54 358 4676232.
E-mail address: vrivarola@exa.unrc.edu.ar (V.A. Rivarola).
1
Both these authors contributed equally to this work.

1357-2725/$ – see front matter © 2008 Elsevier Ltd. All rights reserved.
doi:10.1016/j.biocel.2008.02.024
N.B.R. Vittar et al. / The International Journal of Biochemistry & Cell Biology 40 (2008) 2192–2205 2193

1. Introduction in mitochondria, in the plasma membrane, in endoplasmic


reticulum (ER), Golgi apparatus and lysosomes. It has been
Photodynamic therapy (PDT) involves the combination proposed that intracellular localization is dependent upon
of non-toxic dyes known as photosensitizers (PSs) and the chemical properties of the PS and is cell-line depen-
harmless visible light which in the presence of oxygen leads dent. Localization of PSs in cytoplasmic organelles during
to the generation of reactive oxygen species that can dam- photochemical therapy plays a major role in cell damage.
age cellular constituents leading to cell death. The use of For example, the PSs lutetium texaphyrin (LuTex) and N-
PDT as a cancer therapy is particularly attractive because of aspartyl chlorine e6 (NPe6) cause an almost immediate
its intrinsic dual selectivity. The PS localizes in the malig- disruption of lysosomes upon irradiation (Dougherty et al.,
nant tissue and the light is also spatially focused on the 1998; Agarwal et al., 1991; Oleinick et al., 2002; Woodburn
lesion (Castano, Demidova, & Hamblin, 2005a,b). Provided et al., 1997). In contrast, irradiation of cells preloaded with
that the PS is non-toxic, only the irradiated areas will be the PSs 9-capronyloxy-tetrakis(methoxyethyl) porphycene
affected, even if the PS does bind to normal tissues. (CPO), hexyl pheophorbide (HPPH) and phthalocyanine
Precisely why cells die when subjected to the PDT- (Pc4) has no effect on lysosomes. Instead, one sees a rapid
generated reactive oxygen species (ROS) has been the loss of mitochondrial membrane potential and release of
subject of intense investigation in recent years. The dis- Cytochrome c (Chiu & Oleinick, 2001; Dougherty et al.,
covery of programmed cell death or apoptosis (Samali, 1998; Kessel, Luo, Deng, & Chang, 1997; Kessel, Caruso,
Gorman, & Cotter, 1996) has revolutionized the field of cyto- & Reiners, 2000; Kessel, Luo, Mathieu, & Reiners, 2000;
toxic therapies in general (Dixon, Soriano, Lush, Borner, Oleinick et al., 2002). These effects most likely reflect the
& Figg, 1997) and PDT in particular (Oleinick, Morris, & site of sensitizer accumulation (Trivedi, Wang, Nieminen,
Belichenko, 2002). Oleinick, & Izatt, 2000; Woodburn et al., 1997) because
Agarwal et al. were the first to report apoptosis after the ROS formed upon irradiation have a limited ability to
PDT with chloroaluminium phthalocyanine in mouse lym- migrate from the site(s) of formation (Trivedi et al., 2000).
phoma L5178Y cells. The crucial factors in determining the Apoptosis is an intrinsic physiological event, which
type of cell death, e.g. apoptosis or necrosis following PDT can also be triggered by external stimuli like oxidative
are: cell type, the subcellular localization of the PS, and stress attributable to photosensitization (Castano et al.,
the light dose applied to activate it locally (Agarwal et 2005a). During apoptosis the cells shrink, the nuclear chro-
al., 1991; Alvarez, Prucca, Milanesio, Durantini, & Rivarola, matin becomes pyknotic and condenses against the nuclear
2006; Plaetzer, Kiesslich, Krammer, & Hammerl, 2002). membrane, and eventually the cytoplasm and the nucleus
Phthalocyanines (Pcs) are a new generation of PSs for break up into apoptotic bodies. Although the cytoplasmic
PDT. This group of compounds has several advantages organelles remain intact, DNA is digested at internucleo-
over the sensitizers currently used, which are based on somal sites, giving rise to fragments that are multiples of
hematoporphyrin (Gomes, Cruz, Lopes, Carvalho, & Duarte, 180–200 bp (Almeida, Manadas, Carvalho, & Duarte, 2004).
1999). These azoporphyrins derivatives have stronger An apoptotic response was observed in a variety of tumor
absorbances at longer wavelengths than do porphyrins and cell lines after photosensitization with different porphyrin
tend to have improved photophysical and photochemical and Pc derivatives (He, Skies, Thomsen, Chung, & Jacques,
properties. Their chemical synthesis and structure per- 1994; Kessel & Luo, 1999). However, the ROS that are pro-
mit the addition of substituents to both the central metal duced during PDT have been shown to destroy tumors by
ion and the periphery of the hydrophobic Pc macrocyle, multifactorial mechanisms (Castano et al., 2005a,b). PDT
altering the physical properties of these PSs such as pho- has a direct effect on cancer cells, producing cell death by
tostability, long lifetimes of the photoexcited triplet state, necrosis, apoptosis and autophagy (Oleinick et al., 2002;
and high molar absorption in the red region of the visible Kessel et al., 2006). For most sensitizers in clinical and pre-
spectrum. clinical use, three primary mechanisms of PDT-mediated
In complex environments, such as cells and tissues, the tumor destruction in vivo have been proposed: cellular,
subcellular localization of the PS is important for effective vascular, and immunologic.
photochemistry to occur. Therefore, the cellular structure The aim of the present study is to evaluate the photo-
close to both a high concentration of sensitizer and a high dynamic efficacy of a novel sulfur linked octaalkylamino
oxygen concentration will be preferentially damaged upon substituted zinc(II) phthalocyanine for suitable PDT appli-
illumination. However, many efficient PSs are hydropho- cations. A preliminary study of zinc 1,4,8,11,15,18,22,25-
bic in nature and lose their efficacy when brought into octaalkylphthalocyanines has proven that these com-
the aqueous phase, due to the formation of photochem- pounds are effective PSs with an unexpected mode of PDT
ically inactive aggregates. Amphiphilic carriers, such as action (Cook, Chambrier, Cracknell, Mayes, & Russell, 1995).
micelles (Röder et al., 1996) and liposomes (Röder, Hanke, In this work, we assessed the different Pc S1 delivery
Oelckers, Hackbarth, & Symietz, 2000) are currently used methods with the purpose of disaggregating the PS and cor-
to bring insoluble or hardly soluble dyes into water and relating such parameters with time courses for cell uptake
to avoid aggregation. In these systems, the PS is located using MCF-7c3 human breast cancer cells as an experimen-
in the hydrophobic core of the microheterogeneous host. tal model. To further elucidate the cellular mechanism of
Naturally, suitable carriers should facilitate dye transport action, we examined intracellular localization, lysosomal
without substantially affecting photochemical activity. and mitochondrial changes, with the goal of determining
Many PSs, which are currently used in clinical and exper- the primary photodamage site, which reflects the localiza-
imental treatment of cancer localize extranuclearly in cells: tion of the sensitizer at that time of light irradiation. In
2194 N.B.R. Vittar et al. / The International Journal of Biochemistry & Cell Biology 40 (2008) 2192–2205

addition, we extended our studies to determine whether 1482; 1459; 1405; 1371; 1333; 1295; 1261; 1202; 1168;
necrosis or apoptosis was the prevailing mechanism of 1150; 1087; 1012; 944; 896; 871; 849; 802; 780; 743;
cell death after treatment. In order to optimize the effi- 730; 699; 547; 491 cm−1 . 1 H NMR (500 MHz, DMSO-d6 ):
ciency of this approach, it was essential for us to define the ı = 2.42 (br, 48 H, CH3 ); 3.20 (br, 16 H, CH2 N); 3.71 (br, 16
pharmacological properties of the photosensitizing agent H, SCH2 ); 8.29 (br, 8 H, ArH); HRMS-MALDI-TOF: m/z [M+ ]
in a suitable animal model (LM2 adenocarcinoma subcu- calcd for C64 H88 N16 S8 Zn 1400.4435; found [M+ ] 1400.4635
taneously implanted Balb/c mice) and tumor regression of and [M+ + H] 1401.4579. UV/Vis (DMF): max (␧) = 705 nm
the neoplastic lesion. (2.0 × 105 M−1 cm−1 ).

2. Material and methods 2.3. Liposome preparation

2.1. Cells and culture conditions For use on cells, S1 was dissolved in tetrahydrofuran
(THF) at concentrations indicated in the experiments. It
The human breast cancer MCF-7 (WS8) cell line trans- was then entrapped in liposomes according to Kremer’s
fected with the pBabepuro retroviral vector encoding injection method (Kremer, Esker, Pathmamanoharan, &
human procaspase-3 cDNA (here referred to as MCF-7c3 Wiersema, 1977). For liposomes of d,l-␣-dipalmitoyl
cells) was derived by Dr. C.J. Froelich (Northwestern Uni- phosphatidylethanolamine containing cholesterol
versity, Evanston, IL). The cells were cultured in RPMI (DPPEc), sensitizer (1 ␮mol), phospholipid (33 ␮mol)
1640 media supplemented with 1% l-glutamine, 80 ␮g/ml and cholesterol (7 ␮mol) were dissolved in ethanol-THF
gentamicine and 10% (v/v) fetal bovine serum (FBS). The binary mixture (400 ␮l, 1:1 (v/v)). For l-␣-dimyristoyl-
cultures were maintained at 37 ◦ C in a humidified atmo- phosphatidylcholine liposomes (DMPC), the phospholipid
sphere of 5% CO2 –95% air. (SIGMA) was dissolved in ethanol at a final concentration
of 3 × 10−3 M. About 400 ␮l of the ethanolic solution was
2.2. Photosensitizer then taken up and mixed with 80 ␮l of the photosensitizer
solution. For both liposomes, the resulting solution was
Instrumentation. The 1 H NMR of S1 was recorded on quickly injected through a Hamilton syringe into 5 ml
a Bruker AM 500 and HR-MALDI-TOF mass spectra was of vigorously stirred phosphate-buffered saline (PBS)
measured with a Mod 4700 Applied Biosystems spec- at 80 ◦ C (for DPPEc) or 35 ◦ C (DMPC) depending of the
trometer. Electronic absorption spectra were determined phospholipids employed. The injection was performed at
with a Shimadzu UV-3101 PC spectrophotometer. Fluo- a speed of 50 ␮l/min with magnetic stirring and the final
rescence spectra were monitored with a QuantaMaster volume was reduced to 5 ml by evaporation of organic
Model QM-1 PTI spectrofluorometer. Infrared spectra were solvent. After adding the phthalocyanine stock solution,
performed with a PerkinElmer Spectrum One FT-IR spec- samples were incubated for at least 20 min to allow dye
trometer. incorporation into the liposomes. The preparation was
Synthesis of 2,3,9,10,16,17,23,24-octakis[(N,N-dimethyl- used within a week after preparation.
amino)ethylsulfanyl]phthlocyaninatozinc(II) (referred
here as S1). This compound was readily prepared by the 2.4. Spectroscopic studies
cyclotetramerisation of 1,2-dicyano-4,5-di[(N,N-dimethyl-
amino)ethylsulfanyl]benzene, sinthesized by a method The fluorescence emission spectra of S1 in solutions and
similar to that described in the literature (Gürsoy, Cihan, in cell suspensions were measured using a SPEX Fluoro-
Kocak, & Bekaroglu, 2001). Max spectrofluorometer. The sensitizer fluorescence was
S1 was obtained as described in the above reference collected at the maxima for the S1 excitation and emission
with some minor modifications (Strassert, 2006). at 630 and 713 nm, respectively. The fluorescence signal
A mixture of 1,2-dicyano-4,5-di[(N,N-dimethylamino) was analyzed using the software provided by the manu-
ethylsulfanyl]benzene (0.100 g, 0.30 mmol), anhydrous zinc facturer. All the measurements were performed at room
acetate (0.100 g, 0.50 mmol), and DBU (1 ml, 6.7 mmol) temperature.
in anhydrous butanol (10 ml) was stirred and heated at
130 ◦ C under argon during 2 h. The mixture was cooled 2.5. Quantification of photosensitizer uptake
down, evaporated in vacuo and 5 mL of methylene chlo-
ride was added, and centrifuged to eliminate the excess of Cells were incubated with 1 ␮M of either S1-DPPEc
zinc acetate. The methylene chloride solution was washed or S1-DMPC during a time lapse of 3, 6, 18 and 24 h at
with water (3 × 5 mL) and evaporated in vacuo. The green 37 ◦ C in the dark. After incubation the cell pellets were
solid residue was dissolved in a small volume of toluene collected by scrapping and washed, then dispersed in
and filtered through an aluminium oxide column packed 2 ml of sodium dodecyl sulfate (SDS, in 1:10 water dilu-
and pre-washed with toluene. After washing with toluene tion, v/v). The extraction of sensitizer material from the
and methylene chloride, the title compound was eluted cells was facilitated by agitation. Drug levels in cell pellet
with methanol-methylene chloride (9:1). After evapora- were assessed by fluorescence (excitation 630 nm, emis-
tion in vacuo the dye was recrystallized dissolving in a sion 713 nm) and each value obtained was related to
small volume of toluene–methylene chloride (9:1) follow- the total number of cells contained in the suspension.
ing by the addition of hexane. Yield 0.050 g (50%). IR (KBr): Sensitizer concentration in the samples was calculated
2936; 2858; 2818; 2774; 2572; 2295; 1719; 1623; 1593; from standard calibration curves obtained with known
N.B.R. Vittar et al. / The International Journal of Biochemistry & Cell Biology 40 (2008) 2192–2205 2195

concentrations of S1 treated with SDS. The kinetics of 2.8. Detection of apoptotic cells
uptake (concentration of drug/106 cells vs time) was cal-
culated. Cells grown on coverslips were fixed in PBS contain-
ing 3.7% formaldehyde. Cellular DNA was stained with
1–5 mg/ml Hoechst 33258 and examined by fluorescence
2.6. Photodynamic treatment and viability determination
microscopy. Changes in nuclear morphology were exam-
ined after PDT, by labeling cells with HO258. Apoptotic
One day before the experiment, MCF-7c3 cells
and necrotic cells were identified by characteristic fea-
(2 × 105 cells per dish) were seeded into 35 mm tissue cul-
tures of their nuclei. At least 200 cells were counted from
ture plates in 2 ml RPMI containing 10% FBS, and incubated
each sample, and the yield of apoptotic cells was expressed
overnight (37 ◦ C, 5% CO2 ). On the day of the experiment, the
as the percentage of the total population death. Since
Pc S1 entrapped in DMPC liposomes dispersed into culture
detached cells, which are enriched in apoptotic cells, were
medium supplemented with 4% FBS, was added to cells.
not included in this measurement, the estimated percent-
Culture dishes were placed for 18 h at 37 ◦ C in a cell culture
age of apoptosis determined here is a minimal estimation
incubator. In preparation for irradiation of cells, the dye
of the true apoptosis levels. For the observations of the cel-
containing medium was removed and replaced with RPMI
lular alterations occasioned after treatment, the MCF-7c3
dye-free (10% FBS). For all illuminations, the light source
tumor cells were stained with toluidine blue and visual-
used was a slide projector with a 150 W halogen lamp
ized under light microscope. Fluorescence of HO258 dye
(Reflecta Classic AF-IR) equipped with a water filter with
was imaged using BP 365, FT 395 filters (Carl Zeiss, Ger-
5 cm used to attenuated IR radiation and a cut-off filter
many).
for remove emission wavelengths shorter than 480 nm.
The light intensity at the treatment site was 31 mW cm−2
(Radiometer Laser Mate-Q, Coherent). All irradiations 2.9. Animals and tumor model
were performed at room temperature. After incubation
for 24 additional hours, cell viability was then determined Female Balb/c mice from 6 to 8 weeks old were obtained
by quantization of the cleavage of the tetrazolium salt from the Fundación Balseiro (Buenos Aires) and main-
MTT (3-(4,5-dimethylthiazolil-2)-2,5-diphenyltetrazolium tained in standard cages with free access to tap water and
bromide (SIGMA) by mitochondrial deshydrogenases, normal dietary chow. For the generation of the experimen-
as recommended by the manufacturer (Denizot & Lang, tal tumors, LM-2 cell line (obtained from Hospital Roffo,
1986). Optical density of the resulting solution of formazan Buenos Aires, Argentina) was implanted subcutaneously
salt was read at 540 nm, after subtraction of the blank. by sterile injection of 106 cells in phosphate-buffered
Irradiation of the cells in the absence of Pc did not result in saline (PBS, pH 7.0) into the right forward leg. When
alteration of cell viability (data not shown). Control cells the tumor size reached 0.7 cm on outer diameter, the
were treated in the same way as above without irradiation. tumoral propagation was carried out extracting 2 mm of
Results are presented as percentage of survival, taking tumoral tissue and subcutaneously reimplanted into the
control as 100%. right forward leg of the desired number of mice for each
experiment. The pharmacokinetic and/or phototherapeutic
experiments were performed on the 6th day after implan-
2.7. Subcellular localization and fluorescence staining
tation when tumors reached 0.6–0.7 cm on outer diameter.
procedures
Spontaneous necrosis was minimal or absent for these
tumor sizes.
Tumor MCF-7c3 cells grown on coverslips were incu-
When required, animals were anaesthetized with
bated with 1 ␮M of Pc for 3 h at 37 ◦ C. Then the subcellular
Ketamine (150 mg/kg, by intraperitoneal injection). The
distribution of S1-DMPC was visualized under fluorescence
mice were treated according to the guidelines established
microscopy using a DBP 406/23 + 530/45, DFT 435 + 570,
by the ANMAT Disposition N 6344/96, pp. 1–7 for human
DBP 467/30 + 618/75 filter (Carl Zeiss, Germany).
care of experimental animals.
All fluorescent dyes used in the fluorimetric studies
were purchased from Molecular Probes (Eugene, OR, USA).
Just before imaging, cells were stained with the fluorescent 2.10. Dark toxicity and histopathology examination
dyes by incubation in dye-containing growth medium at
37 ◦ C in the following conditions: LysoTracker® Green DND- S1 into DMPC liposome (0.1, 0.2 and 0.4 mg/kg bw)
26 (75 nM, 15–30 min), used as a probe for check lysosomal was administered by intraperitoneal injection. The animals
localization and photodamage of the Pc, and MitoTracker® were placed in metabolic cages in the dark and six mice
Green FM (100 nM, 45 min), used to study changes in were sacrificed after 7 days S1 administration for histo-
the mitochondria after PDT. Microscopic observations and logical examination. Internal organs such as liver, kidney
photographs were performed using a fluorescence micro- and spleen were excised and fixed in 4% formaldehyde, and
scope (Axiophot, Carl Zeiss, Germany) equipped with a HBO then embedded in paraffin. The blocks were cut 3 ␮m thick
100 W mercury lamp, AxioCam HRc (Carl Zeiss, Germany) and stained with hematoxylin-eosin (H&E) for microscopic
camera and AxioVision Rel. 4.3 software. Images of green analysis.
LysoTracker and MitoTracker fluorescence were collected The histopathological analysis was carried out in the
using a DBP 450–490, FT 510, LP 515 filter (Carl Zeiss, Ger- Animal Pathology Department of Agronomic and Veteri-
many). nary Faculty, UNRC, under PhD Silvia Romanini supervision.
2196 N.B.R. Vittar et al. / The International Journal of Biochemistry & Cell Biology 40 (2008) 2192–2205

2.11. Phototherapeutic studies 3. Results

After tumors reached ∼0.6–0.7 cm in size, the animals 3.1. Dissociation and uptake of S1 phthalocyanine in
were randomized into four groups to determine tumor MCF-7c3 cells
growth rate after the following treatments: (a) control,
not treatment, (b) 0.2 mg/kg bw S1-DMPC, (c) light reg- The chemical structure of S1 and the fluorescence spec-
imen alone: one diary dose of 108 J cm−2 , 600–800 nm, tra in various media are shown in Figs. 1 and 2, respectively.
60 mW cm−2 during three consecutive days (Visona et Fluorescence emission was recorded above 650 nm using a
al., 2000); previous experiments made in our laboratory) low-cut filter. The excitation wavelength was set at 630 nm.
and (d) PDT protocol: 0.2 mg/kg bw and light treatment In solutions, the monomer and aggregated forms can be
24 h dye administration. S1 was given by i.p. injection clearly identified by their band positioned at 715 nm. The
and 24 h later a 1 cm diameter area encompassing the spectra of S1 in DMF solution such as vehiculized in DMPC
tumor was irradiated with the light regime mentioned and DPPEc liposomes are identical. They exhibit a peak at
above from slide projector. The light regimen administrated 715 nm, characteristic of the monomer form. However, the
was chosen based on previous S1 tumor accumulation fluorescence spectrum of S1 in culture medium indicates
assays. We found that 24 h after dye administration a significant contribution by aggregated forms. From the
the tumor present the larger amount of S1 (data not fluorescence spectra results, it seems that aggregation of
shown). S1 can be prevented by an appropriate vehicle. Because
Growth curves representing tumor regrowth for the the aggregation phenomenon is counter productive for
control and treated groups were estimated by measur- effective photodynamic action, we decided to continue our
ing tumors in three dimensions using caliper. Tumor studies evaluating the liposomes as carriers which have
volume (V) was determined by the following equation: shown the drug in disaggregated form.
V = (L × W × H) × 0.5636, where L is length, W is the width Uptake of S1 liposome formulations by MCF-7c3 tumor
and H is the height of the tumor (Whitacre et al., 2000; cells was quantitatively analyzed by fluorescence, a method
Jori, 1995). No spontaneous regression of the tumor was for the rapid measurement of cell-associated dye. MCF-
observed during our investigations. 7c3 cells were incubated with 1 ␮M of phthalocyanine at
37 ◦ C in the presence of 4 and 10% serum in incubation
media. After 3, 6, 18, and 24 h of incubation, phthalocya-
2.12. Evans blue assay for viability determination
nine concentration was measured. The results reported in
Fig. 3A and B demonstrated that S1 formulations entered
With the aim to determinate phototherapeutic effect
the cells in a time-dependent manner. Moreover, the S1-
of S1, tumor necrosis degree was registered following
loaded DMPC liposomes were more rapidly internalized
Schastak et al. (2005). For this, vital stain was performed
to a greater extent than with the S1- DPPEc vehicle. They
by intraperitoneal injection of 0.4 ml 1% Evans Blue (EB)
exhibited different uptake kinetics: S1-DPPEc displayed a
solution. It was injected in the mice after 11 days that
maximum at 18 h (2.2 nmol/106 cells) whereas S1-DMPC
received different treatments: (a) control, not treatment,
accumulated the most within MCF-7c3 cells, reaching the
(b) 0.2 mg/kg bw S1, (c) light regimen alone, and (d) PDT
highest point after 6 h incubation (35 nmol/106 cells). The
protocol as described above. Six hours after administra-
fluorescence intensities measured on MCF-7c3 cells treated
tion of the vital stain, animals were sacrificed, tumors were
excised, and 2–3 mm thick cross-section slices were cut. A
section from the central area of each tumor was examined
macroscopically and photographed using a magnification
of 4×, and analyzed with a PC ImageJ 1.36b program (Wayne
Rasband, National Institute of Health, USA). The unstained
area was attributed to necrotic tissue, whereas the stained
area showed tissue with preserved blood supply. Also, his-
tological cut of tumors were realized as was described
above.

2.13. Statistics

All experiments were repeated at least three times inde-


pendently. Data is shown as the mean and the errors on
graphs represent one standard deviation for at least three
independent values.
Statistical analysis for in vivo experiments were per-
formed using two-way MANOVA’s test (factor 1: treatment,
animal control and treated with S1; factor 2: time). The
DUNCAN test was used to analyze the differences between
the experimental groups. It was considered significant at Fig. 1. Chemical structure of 2,3,9,10,16,17,23,24-octakis[(N,N-
P = 0.05. dimethylamino)ethylsulfanyl]phthlocyaninatozinc(II) (S1).
N.B.R. Vittar et al. / The International Journal of Biochemistry & Cell Biology 40 (2008) 2192–2205 2197

Fig. 2. Absorbance, fluorescence and emission measurements of S1 in solutions and in cell culture medium. (A) Absorption, excitation and emission spectra of
S1 in DMF solution. (B) Comparison of S1 (1 ␮M) emission spectra in DMF solution (solid squares) and; S1-DMPC liposome (open squares), S1-DPPEc liposome
(solid spheres), and S1 (open spheres) in RPMI medium supplemented with 4% FBS. The spectra were recorded on a SPEX FluoroMax spectroflorometer. The
excitation wavelength was set at 630 nm. DMPC: l-␣-dimyristoyl-phosphatidylcholine, DPPEc: d,l-␣-dipalmitoyl phosphatidylethanolamine containing
cholesterol. DMF: dimethylformamide.

with S1-loaded DMPC formulations were at least twenty- 3.2. Photocytotoxicity of S1


fold higher than those obtained with DPPEc by 24 h, at
equivalent dye concentrations. In all cases we observed Photodynamic activity of the S1 was determined on
that incubation with 10% serum, instead of 4% serum, low- MCF-7c3 cells after overnight incubation with 0.1, 0.5 and
ered S1 uptake. Similar was reported by Valduga et al. 1 ␮M followed by 27 J cm−2 dosage of light. Fig. 4A repre-
(1996). Since factors that promote uptake are important sents inhibition of the cell growth (%) plotted against the
(lipophilicity, absence of aggregation and the presence of drug concentration. Using the MTT assay to measure cell
4% serum), S1 incorporated into DMPC liposomes displayed growth at 24 h after photoirradiation, we show that treat-
the most advantageous condition for loading MCF-7c3 ment resulted in 50% cell death (LD50 ) at ∼0.07 ␮M (value
cells and this liposome formulation was chosen for further obtained by extrapolation) and more than 90% cell death at
experiments. concentrations ranging from 0.5 to 1 ␮M (LD100 ). As shown
Our results are in agreement with Ishikawa and Wohrl in Fig. 4B, S1 leads to photokilling of the cells with dif-
who show that phthalocyanines easily aggregate in water, ferent efficacies indicating that cell photoinactivation was
while aggregated phthalocyanine dissociates in hydropho- dependent of both the Pc concentration and the light doses
bic surroundings (Ishikawa, Ohno, Kaizu, & Kobayashi, applied. The different concentrations gave different levels
1992; Wohrle et al., 1994). Similar to this, observed cel- of killing or inhibition of function of MCF-7c3 cells as shown
lular uptake of porphyrins and phthalocyanines has been by their half lethal dose (LD50 ) at 0.5 and 1 ␮M with light
increased with lipophilicity (Berg, Bommer, & Moan, 1989; doses of 17.8 and 8.8 J cm−2 , respectively. Cell survival was
Moan, Berg, Steen, Warloe, & Madslien, 1992; Paquette, Ali, not affected when cultures were maintained in the dark
Langlois, & van Lier, 1988). with the S1 concentrations mentioned above.

Fig. 3. Cellular uptake of S1 in MCF-7c3 cells as a function of incubation time. Cells were incubated for the period of time indicated in the abscissa in
the presence of S1 (1 ␮M) under dark conditions. (A) Cells were incubated with S1 into DPPEc liposome and (B) DMPC. The amount of photosensitizer
incorporated by cells is determined by fluorescence measurements on the extracts as described in the text. Values represent mean ± S.D. of three separated
experiments. (Solid squares: culture medium containing 4% FBS; solid spheres: culture medium containing 10%.)
2198 N.B.R. Vittar et al. / The International Journal of Biochemistry & Cell Biology 40 (2008) 2192–2205

Fig. 4. Effect of S1 photoactivation on MFC-7c3 cell survival. (A) Cells were incubated for 18 h in the presence of different amounts of S1and finally irradiated
(27 J cm−2 ). (B) Cells were pre-treated with S1 (0.5 and 1 ␮M) for 18 h and then irradiated with various light dose. MTT cytotoxicity assay was carried out
24 h after PDT. Results are expressed as the mean ± S.D. of three replicates of each treatment.

3.3. Intracellular localization and sites of photodamage typical mitochondrial-labeling pattern indicates that S1
clearly does not exhibit mitochondrial localization but
The subcellular localization of S1 was investigated by photodynamic treatment using S1 induced mitochondrial
fluorescence microscopy, using MCF-7c3 cells. Fig. 5 shows alternations.
that cells incubated 3 h with 1 ␮M of S1 formulation exhib-
ited red fluorescence typical of ZnPc derivatives and mainly 3.4. Mode of cell death
localized in a cytoplasmic region. In order to determine
whether the staining pattern represented lysosomes, we We next monitored the MCF-7c3 cells to elucidate the
attempted to colocalize S1 with the organelle-specific flu- type of cell death induced by PDT treatment. Analyses
orescent probe LysoTracker Green® (Fig. 5). LysoTracker of morphology and nuclear condensation by HO258 indi-
Green® also exhibited the expected punctuated cytoplas- cate that some of the cells in irradiated cultures preloaded
mic distribution in MCF-7c3 cultures. The merged stained with S1-DMPC were undergoing apoptosis by within 8 h
images revealed a partial overlap of S1 and LysoTracker of irradiation. The percentage of cells displaying nuclear
signals (yellow signal) suggesting that lysosomes are sites chromatin condensation suggestive of apoptosis is shown
of intracellular distribution of S1 dye, similar results were in Fig. 7A. The cells with the mentioned features were
published previously (Bonneau, Morliere, & Brault, 2004; increased by more than 60% after photodynamic treatment
Reiners et al., 2002). The column graph also indicated with 1 ␮M S1 and 8.8 J cm−2 , corresponding to LD50. Cells
the percentage of overlap as spots color quantification treated with S1, but not exposed to light or without S1
(red, green, and yellow) from the merged pictures which but exposed to light, maintained an unaltered appearance.
one revealed more than 70% yellow spots scored corre- Cells began to display nuclear condensation and fragmen-
sponding to signal of co-localization S1 with lysosome tation as indicated by the fluorescent dye at 8 h post-PDT.
probe. The same results were also observed with toluidine blue
When S1 was combined with 27 J cm−2 of visible light, staining including cell rounding, shrinkage and membrane
an immediate phototoxic effect was noted 1 h after PDT at blebbing (Fig. 7).
the level of the mentioned organelle, leading to the loss of
the punctuated LysoTracker Green® staining pattern, and 3.5. S1 in animals: dark toxicity and histopathology
this effect was maintained for the first 3 h post-PDT (Fig. 6). examination
S1 in combination with visible light, but not S1 or light
alone, caused lysosome pattern dye loss, indicating perme- With the purpose of determining the non-toxic PS dose
abilization of lysosomal membranes. for further phototherapeutic treatment, the cytotoxic effect
We next investigated whether PDT induces mitochon- caused by S1 on organs (liver, kidney and spleen) was stud-
drial morphology changes. MitoTracker Green® is taken up ied by histological examinations. For this reason, animals
by the polarized mitochondria. Once inside mitochondria, were treated with different concentrations of S1, and then
MitoTracker Green® binds covalently to sulfhydryls and were sacrificed and the above-mentioned organs were ana-
is retained by mitochondria after depolarization. There- lyzed.
fore, MitoTracker Green® is a suitable dye to monitor Microscopic analyses of liver taken from mice adminis-
mitochondrial volume changes under conditions that pro- tered with 0.1 and 0.2 mg/kg S1 did not show any critical
duce mitochondrial depolarization. As shown in Fig. 6, effect on the normal morphology of the organ (Fig. 8A
before PDT MitoTracker Green® -labeled mitochondria were and B). The biggest concentration evaluated (0.4 mg/kg
bright fluorescent spheres and rods. By 3 h after PDT, mito- S1) leads to hepatocites hypertrophy, degenerative dis-
chondrial morphology changed; mitochondria began to turbances (general hydropic degeneration) and loss of
round and increase in diameter. An examination of the lobulillar structure, as well as travecular structure. Indeed,
N.B.R. Vittar et al. / The International Journal of Biochemistry & Cell Biology 40 (2008) 2192–2205 2199

Fig. 5. Intracellular localization of 1 ␮M of S1 in MCF-7c3 cells after incubation for 3 h. The intracellular distribution and co-localization of S1 with lysosome
were assessed by fluorescence microscopy. MCF-7c3 cells were incubated with both S1 and Lyso Tracker Green DND-26 (75 nM, 15–30 min). Red fluorescence
corresponds to S1 while green fluorescence represents the signal from Lyso Tracker Green DND-26. Yellow fluorescence indicates regions of S1 and Lyso
Tracker Green DND-26 signal overlap. Arrows indicate co-localization spots. Images are representative of those obtained in 3 independent experiments.
Scale bar: 20 ␮m. The column graph represents the percentage of S1 + Lyso Tracker Green signal from merged picture. At least 200 cells were scored. (For
interpretation of the references to color in this figure legend, the reader is referred to the web version of the article.)

hypertrophy of Von Kupffer cells and minor indication of These results allowed us to selection the therapeutic
necrosis was observed (Fig. 8C). concentration of 0.2 mg/kg for further photodynamic treat-
Sections of the kidney stained with H&E showed that ment.
neither 0.1 nor 0.2 mg/kg of S1-DMPC caused any distur-
bance in the physiological morphology of this organ (Fig. 8D 3.6. Phototherapeutic studies
and E). However, a dose of 0.4 mg/kg led to a low degree of
tumefaction from epithelial cells of the proximal and distal The tumor size was measured again 11 days after
contorneated tubes (Fig. 8F). the mice were treated. Tumors that received either no
At the concentrations employed (0.1 and 0.2 mg/kg), S1- treatment, S1 alone or light alone increased their size
DMPC caused no detectable alterations on the structural during the period of observation and there were no sta-
properties of tissue from the spleen (Fig. 8G and H). tistically significant differences between these groups.
On the contrary, spleen from S1-treated animals In contrast, treatment with S1-PDT resulted in partial
(0.4 mg/kg) revealed large degeneration and cell death tumor regression in all tumors within 10 days after PDT.
areas (Fig. 8I). Measurement of the tumor volume during the treat-
2200 N.B.R. Vittar et al. / The International Journal of Biochemistry & Cell Biology 40 (2008) 2192–2205

Fig. 6. S1-PDT causes loss of lysosomal integrity and mitochondrial swelling. Cells grown on coverslips were exposed to 75 mM of LysoTraker Green DND-26
during 30 min (upper panel) or 100 mM of Mito Traker Green FM during 45 min (lower panel). After that, the cells were treated with S1 1 ␮M alone or with
S1 1 ␮M and 27J cm−2 . Images were collected 3 h after PDT treatment. Scale bar: 10 ␮m.

ment revealed a growth delay of ∼5–6 days in tumor 3.7. Tumor viability assay and histological analysis of
growth in the S1-PDT treated mice. Estimates of contrast S1-PDT treated-tumors
between S1-PDT and each of the other three treatments
mentioned above were statistically different (p < 0.05) In the tumors of the untreated, light and S1-
(Fig. 9). dark groups, a total staining with Evans Blue was

Fig. 7. PDT induces apoptosis in MCF-7c3 cells. (A) Percentage of cell death from PDT-treated cultures 8 h later (1 (M S1 and 8.8 J cm−2 , corresponding
to LD50). (B) Upper panel, fluorescence photomicroghaphs of MCF-7c3 cells stained with Hoechst 33258: (left) untreated; (right) MCF-7c3 cells 8 h after
treatment with LD50. Arrows indicate apoptotic nuclei. Lower panel, micrographs of MCF-7c3 cells stained with TB: (left) untreated; (right) cells after 8 h
PDT treatment. Arrows indicate plasma membrane blebbing. Scale bar: 10 ␮m. TB: toluidine blue dye.
N.B.R. Vittar et al. / The International Journal of Biochemistry & Cell Biology 40 (2008) 2192–2205 2201

Fig. 8. Histological examinations on different organs from mice treated with several concentrations of S1. Microscopic appearance of liver (A–C), kidney
(D–F), and spleen (G–I) obtained at 7 days from mice treated with S1: (A, D and G) 0.1 mg kg−1 ; (B, E, and H) 0.2 mg kg−1 ; (C, F, and I) 0.4 mg kg−1 (×400).
The specimen was stained with H&E. The discontinuous circles on the photograph are representatives of the histopathological alterations described in the
text.

observed (Fig. 10), which indicates that death in In the tumors of the control group the histological cut
those tumors does not occur. Mice which received showed evidences of malignancy, such as imprecise mar-
Evans Blue after PDT, showed an increased tumor gins with infiltrative growth in muscle, mitotic figures,
death. anisokaryosis, pleomorphism and vascularization. Those
characteristics have also been observed for light and S1-
dark groups (data not shown). In PDT-treated tumors, large
quantity of dead tumor cells can be observed. The sections
presented pycnotic nucleus, sign of injury with principles
of necrosis. The evaluated tumor sections did not present
capsule of connective tissue and we observed intratu-
moral hemorrhages, as a consequence of the S1-PDT effects
(Fig. 11).

4. Discussion

A novel zinc(II) phthalocyanine derived photosensi-


tizer was evaluated for the potential use in PDT. In
vitro screening of S1 administration systems efficacy
should be helpful in determining the most satisfactory
Fig. 9. Effect of S1-PDT on tumors. Tumors were either untreated or formulation, which would then merit further in vivo
treated with red light alone, S1alone or S1-PDT (0.2 mg kg−1 and red light). evaluation. Comparison of different formulations in the
The data represent the means ± S.E.M. of the volumes of four tumors in
same system might lead to a better understanding of the
each experimental group. Black arrow indicates the beginning of the treat-
ment and grey arrows indicates the light dose applied. Dosage of light: mechanisms involved in the cellular toxicity of S1 and
108 J cm−2 . p < 0.05 S1-PDT vs untreated, S1 and light. would be useful in defining which parameters should
2202 N.B.R. Vittar et al. / The International Journal of Biochemistry & Cell Biology 40 (2008) 2192–2205

Fig. 10. Evans blue assay on PDT-treated tumors for viability determination. The vital stain was performed by intraperitoneal injection of 0.4 ml 1% EB
solution after 11 days that received different treatments: (A) untreated, (B) light, (C) S1, and (D) S1-PDT. A section from the central area of each tumor was
examined macroscopically and photographed using a magnification of 4×. Unstained area—tumor necrosis, stained area—viable tissue.

be controlled in the preparation of an optimal formula- the length of the saturated carbon chain (14 carbon atoms
tion. in DMPC, 16 in DPPE), the transition temperature (22 ◦ C for
The first part of our study was to determinate that the DMPC and 41 ◦ C for DPPE), and the presence of cholesterol
DMPC-liposome fusion was the best way for delivering the in DPPE.
dye. Previous reports have shown that Pc aggregation is In addition, it is known that the effect of the serum
easily detected spectroscopically due to the fact that aggre- on cellular uptake is counter productive; we observed this
gated Pcs do not fluoresce, Pcs in their monomeric forms do, event when overnight incubation with S1-DMPC was car-
and thus aggregated Pcs do not serve as PSs (Allen, Langlois, ried out with 10% serum, probably molecules are bound to
Sharman, La Madeleine, & Van Lier, 2002; Oda, Ogura, & serum which prevents diffusion and limits cell penetration
Ocurra, 2000). Our data from fluorescent spectra clearly mechanisms by endocytosis. In vitro, serum proteins have
demonstrates that S1 in culture media was as monomer been reported to compete with cells for Pc derivatives, thus
employing liposomal formulations, whereas was found in decreasing cell uptake of the dye (Konan, Chevallier, Gurny,
aggregated form without carriers. & Allemann, 2003; Rodal, Rodal, Moan, & Berg, 1998).
The results of the uptake assays, where this mechanism It is evident that S1-DMPC has photodynamic activity on
has been influenced by the PS aggregation grade, support MCF-7c3 cancer cells. The irradiation regime of sensitized
the above mentioned results. PSs may be taken up by cells cells showed a decrease in cell viability (MTT assay 24 h
either directly through the diffusion-mediated pathway or post irradiation) in a concentration and light dose depen-
by endocytosis (Berg & Moan, 1997). We could gain some dent manner (Fig. 4A). Our results demonstrated that in
insight into the entry process by considering the differences the cultures incubated with S1-DMPC followed by 27 J cm−2
found in the amount of S1 after incorporation employ- irradiation level, there was a loss in viability, leading to a
ing DMPC vesicles. The normal function of these processes ∼70% inactivation at 0.5 ␮M whereas a substantially greater
could be impeded in the case of S1 administered without phototoxic effect was achieved at 1 ␮M S1-DMPC, which
vesicles leading to a possible accumulation of larger aggre- resulted in ∼100% cell death (Fig. 4B).
gates on the plasma membrane that could be inhibiting PS uptake by cancer cells is crucial for effective PDT and
the penetration and subsequent action of the dye. More- therefore, to a certain degree, the type of photodamage that
over, in the case of S1 delivery using DPPEc liposome, we occurs in cells loaded with a PS and illuminated depends
found that the intracellular concentration level of the Pc on the precise of subcellular localization of the PS within
was lower than using DMPC (Fig. 3). The liposomal formu- the cell.
lations used differ in terms of phospholipid composition; In the present study the visualization of subcellular
these differences could explain in part the variability of the localization of S1 was achieved by the use of fluores-
results which have been reported concerning the reduced cence microscopy techniques. The fluorescence localization
uptake of S1-DPPEc liposome by cells. The differences were pattern observed suggest that this agent would be accu-

Fig. 11. Tumor specimen obtained from PDT-treated mice. H&E stained micrographs of PDT-treated tumor 11 days later. PDT dose: 0.2 mg kg−1 bw S1 by i.p.
injection and red light exposure, 108 J cm−2 three consecutive days. The micrograph (×400) shows a large area of cell death (discontinued circles).
N.B.R. Vittar et al. / The International Journal of Biochemistry & Cell Biology 40 (2008) 2192–2205 2203

mulated by lysosomes. Similar results have been reported microscopically tumor tissue after the therapy confirmed
for PS with affinity for this organelle (Castino, Demoz, & cell death by mean of necrosis pathways. These data did not
Isidoro, 2003; Kessel, Caruso et al., 2000; Kessel, Luo et al., reflect our previously in vitro results where the preferential
2000; Reiners et al., 2002; Bonneau et al., 2004). Colocaliza- cell destruction was by apoptosis. The extrapolation of in
tion of subcellular organelle specific probes with differing vitro observations to in vivo mechanisms is not always pos-
fluorescence emission maxima than that of the PS can sible due to the tumor environment differs completely with
be used to more closely identify the site of localization the in vitro conditions. In an interesting approach to exploit-
(Wilson, Olivo, & Singh, 1997) and these probes can also be ing immune effects, Castano and colleagues (Castano, Mroz,
used to identify sites of damage after illumination (Kessel & Hamblin, 2006) reported that PDT kill malignant cells by
et al., 1997). Our findings from fluorescence localization necrosis stimulating the host immune system.
patterns of LysoTracker Green sensor correlated with the The photosensitizing agent used in the present report
photosensitizer thereby demonstrating an affinity for this give an approach that the development of experimental
cellular compartment (Fig. 5). In addition, the fluorescence protocols in vivo can contribute to understanding of the
of lysosomal probe became much more diffuse 1 h after PDT photokilling effect in basic oncological research and to
dose application. This observation clearly places the lyso- assess the potential for clinical applications in PDT of can-
some as a primary target of S1 photoactivation due to the cer.
intracellular localization of oxidative stress generated by
PDT coinciding with the intracellular localization of the PS Acknowledgments
(Fig. 6).
Moreover, using MitoTracker Green sensor, a marked Authors are grateful to Consejo Nacional de Investiga-
effect was noted at the level of mitochondria, showing com- ciones Cientı́ficas y Técnicas (CONICET) Argentina, Agencia
plete alteration of the morphology within the first 3 h after Nacional de Promoción Cientı́fica y Tecnológica and SECYT,
PDT, seen as a mitochondrial swelling (Fig. 6). Under these Universidad Nacional de Rı́o Cuarto for financial support.
PDT conditions, we have found morphologic features of V.R. and J.A. holds a position of Researchers at CONICET.
apoptosis such as cell shrinkage, chromatin condensation, R.V.N.B. thanks CONICET for a research fellowship. P.C.G.
and nuclear fragmentation in MCF-7c3 tumor cells after S1 thanks UNRC for a student fellowship.
treatment (Fig. 7). Similar to our findings, photoactivation The authors thank to Romanini S. for histological exam-
of the phthalocyanine Pc4 was reported to induce mito- inations. R.V., R.V.N.B., and P.C.G. thank to Argenta Price for
chondrial swelling after inner mitochondrial membrane her critical review of the manuscript.
permeabilization and depolarization, resulting in apoptotic
death (Lam, Oleinick, & Nieminen, 2001). References
Apoptosis may be initiated by a wide array of stimuli
that trigger a variety of signaling pathways that, for the Agarwal ML, Clay ME, Harvey EJ, Evans HH, Antunez AR, Oleinick NL. Pho-
todynamic therapy induces rapid cell death by apoptosis in L5178Y
most part, converge at the mitochondrion (Ferri & Kroemer, mouse lymphoma cells. Cancer Research 1991;51:5993–6.
2001). Thus, it has been shown that an apoptotic response Ali SM, Chee SK, Yuen GY, Olivo M. Photodynamic therapy induced Fas-
can be obtained when lysosomes (Berg & Moan, 1994; mediated apoptosis in human carcinoma cells. International Journal
of Molecular Medicine 2002;9:257–70.
Berg & Moan, 1997; Nagata, Obana, Gohto, & Nakajima,
Allen CM, Langlois R, Sharman WM, La Madeleine C, Van Lier
2003; Woodburn et al., 1997) or mitochondria (Ali, Chee, JE. Photodynamic properties of amphiphilic derivatives of alu-
Yuen, & Olivo, 2002; Morgan & Oseroff, 2001; Theodossiou, minum tetrasulfophthalocyanine. Photochemistry & Photobiology
2002;76:208–16.
Noronha-Dutra, & Hothersall, 2006) are targeted for light
Almeida RD, Manadas BJ, Carvalho AP, Duarte CB. Intracellular signaling
induced damage. Furthermore, several reports have related mechanisms in photodynamic therapy. Biochimica et Biophysica Acta
a variety of cytotoxic agents that trigger lysosomal damage- 2004;1704:59–86.
induced apoptosis (Neuzil, Svensson, Weber, Weber, & Alvarez MG, Prucca C, Milanesio ME, Durantini EN, Rivarola V. Photody-
namic activity of a new sensitizer derived from porphyrin-C60 dyad
Brunk, 1999; Roberg, Johansson, & Ollinger, 1999). Given and its biological consequences in a human carcinoma cell line. Inter-
the susceptibility of lysosomes to oxidant-induced injury; it national Journal of Biochemistry & Cell Biolology 2006;38:2092–101.
is conceivable that lysosomal disruption may be a relatively Berg K, Bommer JC, Moan J. Evaluation of sulfonated aluminum phthalo-
cyanines for use in photochemotherapy. Cellular uptake studies.
common initiator of the mitochondrial/intrinsic apoptotic Cancer Letters 1989;44:7–15.
pathway. Berg K, Moan J. Lysosomes as photochemical targets. International Journal
We concluded that S1 had a primary site of accumula- of Cancer 1994;59:814–22.
Berg K, Moan J. Lysosomes and microtubules as targets for pho-
tion and photodamage in lysosomes, and cells underwent tochemotherapy of cancer. Journal of Photochemistry and Photobi-
apoptosis upon illumination doses leading to 60% cell ology 1997;65:403–9.
death, suggesting that apoptotic pathways caused by PDT Bonneau S, Morliere P, Brault D. Dynamics of interactions of photosensitiz-
ers with lipoproteins and membrane-models: correlation with cellular
may be activated via lysosomes followed by mitochondrial
incorporation and subcellular distribution. Biochemical Pharmacol-
destabilization, which leads to amplification of the stim- ogy 2004;68:1443–52.
uli. The same phenomenon was observed in reports with Boya P, Andreau K, Poncet D, Zamzami N, Perfettini JL, Metivier D, et
al. Lysosomal membrane permeabilization induces cell death in a
the chlorin-based PS, ATX-S10(Na) a lysosomally located
mitochondrion-dependent fashion. Journal of Experimental Medicine
photosensitizer (Nagata et al., 2003). 2003;197:1323–34.
Our in vivo studies clearly showed that the treatment Castano AP, Demidova TN, Hamblin MR. Mechanisms in photodynamic
was well tolerated and without any significant side effects therapy: part two-cellular signaling, cell metabolism and modes of
cell death. Photodiagnosis & Photodynamic Therapy 2005a;2:1–23.
as well as its potential tumor regrowth. This regression was Castano AP, Demidova TN, Hamblin MR. Mechanisms in photo-
attributed to the combination of S1 and light. Moreover, dynamic therapy: part three photosensitizer, pharmacokinetics,
2204 N.B.R. Vittar et al. / The International Journal of Biochemistry & Cell Biology 40 (2008) 2192–2205

biodistribution, tumor localization and modes of tumor destruction. Moan J, Berg K, Steen HB, Warloe T, Madslien K. Fluorescence and
Photodiagnosis & Photodynamic Therapy 2005b;2:91–106. photodynamic effects of phthalocyanines and porphyrins in cells.
Castano AP, Mroz P, Hamblin MR. Photodynamic therapy and anti-tumour In: Henderson BW, Dougherty TJ, editors. Photodynamic ther-
immunity. Nature Review Cancer 2006;6:535–45. apy. New York/Basel/Hong Kong: Marcel Dekker Inc.; 1992. p.
Castino R, Demoz M, Isidoro C. Destination “lysosomes”: a target 19–35.
organelle for tumor cell killing? Journal of Molecular Recognition Morgan J, Oseroff AR. Mitochondria-based photodynamic anti-cancer
2003;16:337–48. therapy. Advanced Drug Delivery Reviews 2001;49:71–86.
Chen Y, Mak NK, Yow CMN, Fung MC, Chiu LC, Leung WN, et al. The binding Nagata S, Obana A, Gohto Y, Nakajima S. Necrotic and apoptotic cell death
characteristics and intracellular localization of temoporfin (mTHPC) of human malignant melanoma cells following photodynamic ther-
in myeloid leukemia cells: phototoxicity and mitochondrial damage. apy using an amphiphilic photosensitizer ATX-S10(Na). Lasers Surgery
Photochemistry & Photobiology 2000;72:541–7. Medicine 2003;33:64–70.
Chiu SM, Oleinick NL. Dissociation of mitochondrial depolarization from Neuzil J, Svensson I, Weber T, Weber C, Brunk U. Alpha-tocopheryl
Cytochrome c release during apoptosis induced by photodynamic succinate-induced apoptosis in Jurkat T cells involves caspase-3 acti-
therapy. British Journal of Cancer 2001;84:1099–106. vation, and both lysosomal and mitochondrial destabilization. FEBS
Cook MJ, Chambrier I, Cracknell SJ, Mayes DA, Russell DA. Octa-alkyl Letters 1999;445:295–300.
zinc phthalocyanines: potential photosensitizers for use in the pho- Oda K, Ogura S, Ocurra I. Preparation of a water-soluble fluorinated zinc
todynamic therapy of cancer. Photochemistry and Photobiology phthalocyanine and its effect for photodynamic therapy. Journal of
1995;62(3):542–5. Photochemistry & Photobiology B: Biology 2000;59:20–5.
Denizot F, Lang R. Rapid colorimetric assay for cell growth and Oleinick NL, Morris RL, Belichenko I. Apoptosis in response to pho-
survival. Modifications to the tetrazolium dye procedure giving todynamic therapy: what, where, why and how. Photochemical &
improved sensitivity and reliability. Journal of Immunological Meth- Photobiological Sciences 2002;1:1–21.
ods 1986;89:271–7. Paquette B, Ali H, Langlois R, van Lier JE. Biological activities
Dixon SC, Soriano BJ, Lush RM, Borner MM, Figg WD. Apoptosis: its role in of phthalocyanines-VIII. Cellular distribution in V-79 Chinese
the development of malignancies and its potencial as a novel thera- hamster cells and phototoxicity of selectively sulfonated alu-
peutic target. Annals of Pharmacotherapy 1997;31:76–82. minum phthalocyanines. Photochemistry & Photobiology 1988;47:
Dougherty TJ, Gomer CJ, Henderson BW, Jori G, Kessel D, Korbelik M, et 215–20.
al. Photodynamic therapy. Journal of the National Cancer Institute Plaetzer K, Kiesslich T, Krammer B, Hammerl P. Characterization of the
1998;90:889–905. cell death modes and the associated changes in cellular energy supply
Ferri KF, Kroemer G. Organelle-specific initiation of cell death pathways. in response to AlPcS4-PDT. Photochemical & Photobiological Sciences
Nature Cell Biology 2001;3:E255–63. 2002;1:172–7.
Ginevra F, Biffanti A, Pagnan R, Biolo E, Reddi E, Jori G. Delivery of the tumor Reiners Jr JJ, Caruso JA, Mathieu P, Chelladurai B, Yin X-M, Kessel D. Release
photosensitizer zinc(II)-phthalocyanine to serum proteins by differ- of Cytochrome c and activation of pro-caspase-9 following lysoso-
ent liposomes: studies in vitro and in vivo. Cancer Letters 1990;49: mal photodamage involves Bid cleavage. Cell Death & Differentiation
59–65. 2002;9:934–44.
Gomes ER, Cruz T, Lopes CF, Carvalho AP, Duarte CB. Photosensitization Roberg K, Johansson U, Ollinger K. Lysosomal release of cathepsin D
of lymphoblastoid cells with phthalocyanines at different saturating precedes relocation of Cytochrome c and loss of mitochondrial trans-
incubation times. Cell Biology & Toxicology 1999;15:249–60. membrane potential during apoptosis induced by oxidative stress.
Gürsoy S, Cihan A, Kocak MB, Bekaroglu O. Synthesis of new metal-free Free Radicals Biology Medicine 1999;27:1228–37.
and metal-containing phthalocyanines with tertiary or quaternary Rodal GH, Rodal SK, Moan J, Berg K. Liposome-bound Zn (II)-
aminoethyl substituents. Monatshefte für Chemie 2001;132:813–9. phthalocyanine. Mechanisms for cellular uptake and photosen-
He XY, Skies RA, Thomsen S, Chung LW, Jacques SL. Photodynamic therapy sitization. Journal of Photochemistry & Photobiology, B: Biology
with Photofrin II induces programmed cell death in carcinoma cell 1998;45:150–9.
lines. Photochemistry & Photobiology 1994;59:468–73. Röder B, Hackbarth S, Korth O, Zimmermann C, Herter R, Hanke T, et
Ishikawa N, Ohno O, Kaizu Y, Kobayashi H. Localized orbital study on the al. Photophysical properties of pheophorbide a in different carrier-
electronic structures of phthalocyanine dimmers. Journal of Physical systems. Proc SPIE The International Society for Optical Engineering
Chemistry 1992;96:8832–9. 1996;2625:179–86.
Ishisaka R, Utsumi T, Yabuki M, Kanno T, Furuno T, Inoue M, et al. Activation Röder B, Hanke T, Oelckers S, Hackbarth S, Symietz C. Photophysical
of caspase-3-like protease by digitonintreated lysosomes. FEBS Letters properties of pheophorbide a in solution and in model mem-
1998;435:233–6. brane systems. Journal of Porphyrins & Phthalocyanines 2000;4:
Jori G. Photodynamic therapy: basic and preclinical aspects. In: Horspool 37–44.
WM, Song PS, editors. CRC handbook of organic photochemistry & Samali A, Gorman AM, Cotter TG. Apoptosis: the story so far. Experimentia
photobiology; 1995. p. 1379–83. 1996;52:933–41.
Kessel D, Luo Y, Deng Y, Chang CK. The role of subcellular localization in Schastak S, Jean B, Handzel R, Kostenich G, Hermann R, Sack U,
initiation of apoptosis by photodynamic therapy. Photochemistry & et al. Improved pharmacokinetics, biodistribution and necrosis in
Photobiology 1997;65:422–6. vivo using a new near infra-red photosensitizer: tetrahydropor-
Kessel D, Luo Y. Photodynamic therapy: a mitochondrial inducer of apop- phyrin tetratosylat. Journal of Photochemistry & Photobiology
tosis. Cell Death & Differentiation 1999;6:28–35. 2005;78:203–13.
Kessel D, Caruso JA, Reiners Jr JJ. Potentiation of photodynamic therapy by Strassert, C. A. (2006). Thesis. University of Buenos Aires. Facultad de Far-
ursodeoxycholic acid. Cancer Research 2000a;60:6985–8. macia y Bioquimica, p. 95 [unpublished results].
Kessel D, Luo Y, Mathieu P, Reiners Jr JJ. Determinants of the apoptotic Theodossiou TA, Noronha-Dutra A, Hothersall JS. Mitochondria are a
response to lysosomal photodamage. Photochemistry & Photobiology primary target of hypericin phototoxicity: Synergy of intracellular cal-
2000b;71:196–200. cium mobilisation in cell killing. International Journal of Biochemistry
Kessel D, Graça M, Vicente H, Reiners Jr JJ. Initiation of apoptosis & Cell Biology 2006;38:1946–56.
and autophagy by photodynamic therapy. Lasers Surgery Medicine Trivedi NS, Wang HW, Nieminen AL, Oleinick NL, Izatt JA. Quantita-
2006;38:482–8. tive analysis of Pc 4 localization in mouse lymphoma (LY-R) cells
Konan YN, Chevallier J, Gurny R, Allemann E. Encapsulation of p-THPP into via double-label confocal fluorescence microscopy. Photochemistry &
Nanoparticles: Cellular Uptake, Subcellular Localization and Effect Photobiology 2000;71:634–9.
of Serum on Photodynamic Activity. Photochemistry & Photobiology Valduga G, Bianco G, Csik G, Reddi E, Masiero L, Garbisa S, et al.
2003;77:638–44. Interaction of hydro- lipophilic phthalocyanines with cells of dif-
Kremer JMH, Esker MWJ, Pathmamanoharan C, Wiersema PH. Vesicles of ferent metastatic potential. Biochemical Pharmalology 1996;51:
variable diameter prepared by a modified injection method. Biochem- 585–90.
istry 1977;16:3932–5. Visona A, Angelini A, Gobbo S, Bonanome A, Thiene G, Pagnan A, et al.
Lam M, Oleinick NL, Nieminen AL. Photodynamic therapy-induced apop- Local photodynamic therapy with Zn(II)-phthalocyanine in an exper-
tosis in epidermoid carcinoma cells. Reactive oxygen species and imental model of intimal hyperplasia. Journal of Photochemistry &
mitochondrial inner membrane permeabilization. Journal of Biolog- Photobiology B: Biology 2000;57:94–101.
ical Chemistry 2001;276:47379–86. Whitacre CM, Feyes DK, Satoh T, Grossmann J, Mulvihill JW, Mukhtar H, et
Li W, Yuang X, Nordgren G, Dalen H, Dubowchik GM, Firestone RA, et al. al. Photodynamic therapy with the phthalocyanine photosensitizer Pc
Induction of cell death by the lysosomotropic detergent MSDH. FEBS 4 of SW480 human colon cancer xenografts in athymic mice. Clinical
Letters 2000;470:35–9. Cancer Research 2000;6:2021–7.
N.B.R. Vittar et al. / The International Journal of Biochemistry & Cell Biology 40 (2008) 2192–2205 2205

Wilson BC, Olivo M, Singh G. Subcellular localization of Photofrin cesses of sulfur compounds and in photodynamic therapy of cancer.
and aminolevulinic acid and photodynamic crossresistance in vitro Russian Chemical Bulletin 1994;43:1953–64.
in radiation-induced fibrosarcoma cells sensitive or resistant to Woodburn KW, Fan Q, Miles DR, Kessel D, Luo Y, Young SW. Localiza-
Photofrin-mediated photodynamic therapy. Photochemistry & Pho- tion and efficacy analysis of the phototherapeutic lutetium texaphyrin
tobiology 1997;65:166–76. (PCI-0123) in the murine EMT6 sarcoma model. Photochemistry &
Wohrle D, Wendt A, Weitemeyer A, Stark J, Spiller W, Muller S, et al. Metal Photobiology 1997;65:410–5.
chelates of porphyrin derivatives as sensitizers in photooxidation pro-

You might also like