You are on page 1of 9

Journal of Porphyrins and Phthalocyanines

J. Porphyrins Phthalocyanines 2001; 5: 161–169

Current status of phthalocyanines in the photodynamic


therapy of cancer

CYNTHIA M. ALLEN, WESLEY M. SHARMAN and JOHAN E. VAN LIER*

MRC Group in the Radiation Sciences, Faculty of Medicine, Université de Sherbrooke, Sherbrooke, Québec, Canada J1H 5N4

Received 7 January 2000; Revised 15 March 2000


Accepted 3 April 2000

ABSTRACT: Photodynamic therapy is a binary treatment now accepted in clinic for various malignancies in several
countries around the world. Phthalocyanine molecules are second-generation photosensitizers with enhanced
photophysical and photochemical properties over those of porphyrins. They have been shown to be phototoxic against
a number of cell types and tumor models. A great deal of research has been devoted to the elucidation of their
mechanism of action and mode of cell death. The present paper reviews phthalocyanine pre-clinical anti-cancer
research with emphasis on phthalocyanine induced apoptosis using a silicon phthalocyanine, Pc4. A brief summary of
the latest clinical results using phthalocyanines is presented. Copyright # 2001 John Wiley & Sons, Ltd.

KEYWORDS: apoptosis; photodynamic therapy; phthalocyanines; Pc4; Photofrin1; photosensitizer; clinical trials

INTRODUCTION agents produced during PDT. This induces a two-fold


selectivity, as there is a preferential uptake of the
In North America, cancer is one of the leading causes of photosensitizer by the diseased tissue as well as the capacity
death and disease. In 1999, over 1.2 million new cases of to confine activation of the photosensitizer to the tumor by
malignancies were diagnosed in the United States with restricting the illumination to that specific region. As such,
Canada having similar statistics per capita, 4 cases per 1000 PDT allows for the exclusive eradication of tumor tissue
individuals (Canadian and American Cancer Societies). while sparing surrounding healthy cells from damage.
Traditional cancer therapies such as surgery, chemotherapy The phenomenon of photodynamic action may well date
and radiation therapy involve a delicate balance between as far back as primitive man with the use of plant extracts
removing or destroying diseased tissue and sparing and sunlight to cure various skin afflictions. In India, as
surrounding healthy cells. These conventional treatments early as 1400 B.C., psoralens, obtained from the seeds of
procure serious side effects caused by the loss of normal cell Psoralea corylifolia, were used in the treatment of vitiligo
function due to relatively indiscriminate cytotoxic proper- [1]. Photodynamic action, however, was not formally
ties. The development of new cancer therapies with reported until the turn of the twentieth century when a
improved selectivity is required. medical student, Oscar Raab, working under the direction of
Dr H. von Tappeiner in Munich, noticed the lethal effects of
an acridine orange solution on Paramecia, which were
dependant on the light intensity in the laboratory [2]. This
PHOTODYNAMIC THERAPY phenomenon was more extensively studied by von Tappei-
ner and Jodlbauer [3] who coined the term “photodynamic
A novel cancer treatment that is receiving increased action”. It was noted that other such compounds such as
attention is photodynamic therapy (PDT). PDT is a binary eosin were able to induce rapid cell kill in the presence of
therapy that involves the combination of visible light and a light. It was established that dissolved molecular oxygen is
photosensitizer. Each component is harmless by itself, but, needed for the reaction to occur. Hausmann in 1908 reported
in combination with molecular oxygen, can lead to the the photohemolysis of rabbit erythrocytes using hemato-
generation of reactive oxygen species (ROS), oxidative cell porphyrin (Hp) [4, 5]. Various scientists [6–8] went on to
damage and cell death. The duality of this treatment leads to demonstrate photosensitization of mice, guinea pigs and
greater selectivity towards destroying diseased tissue since man respectively, by inoculation with hematoporphyrin
only cells that are exposed simultaneously to the photo- (Hp). An excellent historical perspective has recently been
sensitizer, light and oxygen are subjected to the cytotoxic published [9].

———————
*Correspondence to: J. E. Van Lier, Department of Nuclear Medicine Mechanisms of Photosensitization
and Radiobiology, Faculty of Medicine, Université de Sherbrooke,
Sherbrooke, Québec, Canada J1H 5N4. Upon illumination, the photosensitizer used in PDT is
E-mail: jvanlier@courrier.usherb.ca excited to its first excited singlet state. This excited state is
far too short-lived to effectively interact with its surround-
Copyright # 2001 John Wiley & Sons, Ltd. ings and rapidly loses its energy via radiative and non-
162 C. M. ALLEN ET AL.

radiative decay. In terms of PDT, the most important of similar short ranges in cellular systems. Furthermore, it has
these is intersystem crossing to populate the much longer been recently shown that Type I and Type II mechanisms
lived triplet state. This excited state usually has a lifetime in probably act in harmony in most cases [20–22]. Overall,
the ms range [10], which is sufficient time for interaction however, the initial reaction, whether via a Type I or a Type
between the excited photosensitizer and surrounding mol- II mechanism, is not of such importance as both lead to
ecules. As such, it is generally accepted that generation of similar oxidative damage and free radical chain reactions.
cytotoxic species during PDT involves interactions with the Ultimately, both mechanisms lead to important oxidative
excited triplet state of the sensitizer, although examples damage within the target tissue, inducing cell death and
exist where the singlet state is involved [11]. tumor destruction.
This excited triplet state can interact in one of two ways,
defined as Type I and Type II. A Type I mechanism involves
a hydrogen atom abstract or electron transfer reaction
leading to the generation of free radicals and radical ions. PHOTOFRIN1
Such species can readily react with molecular oxygen,
inducing irreparable oxidative cellular damage and the The first photosensitizers accepted for clinical use by
production of ROS, which can lead to further biological governmental regulatory agencies are the first-generation
destruction. The role of reactive oxygen species in the hematoporphyrin derivatives such as Photofrin1 (Fig. 1).
photodynamic process has recently been reviewed [12]. On Photofrin1, a mixture enriched in the more active dimeric
the other hand, an energy transfer reaction to molecular and oligomeric components, has been accepted in several
oxygen is defined as a Type II mechanism. The resulting countries for the treatment of early- and late-stage lung
reactive species, singlet oxygen, is a zwitterionic species cancer, superficial and advanced oesophageal cancer,
and reacts rapidly with numerous biologically important bladder cancer, superficial and early-stage gastric cancer,
substrates, again leading to important oxidative damage and early-stage cervical cancer and cervical dysplasia. In
ultimately cell death. It is generally accepted that the Type addition, Photofrin1 is being investigated as a possible
II mechanism predominates during PDT and that singlet therapy for a number of other malignant and non-malignant
oxygen is the most important cytotoxic species produced conditions. Other hematoporphyrin derivatives are also
[12–15]. The range of singlet oxygen in cellular media is being used, with Photoheme, an enriched hematoporphyrin
limited to approximately 45 nm [16, 17]. With the diameter derivative similar to Photofrin1 accepted for clinical use in
of human cells ranging from 10 to 100 mm, the site of Russia for easily accessible cancers including skin, breast,
primary generation of singlet oxygen consequently deter- oropharingeal, lung, larynx and gastrointestinal cancers
mines which subcellular target is attacked, either initiating along with psoriasis and prophylaxis for corneal transplant
an apoptotic or necrotic response. Type I reactions become opacity and recurrent blindness, where newly formed blood
more important at low oxygen concentrations or in polar vessels result in loss of corneal transparency [23–26].
environments [18, 19] and produce reactive oxygen species Despite the clinical success achieved using Photofrin1,
such as superoxide anion and hydroxyl radicals that have this first-generation photosensitizer has several important

Fig. 1. Photofrin1 structure consisting of a complex mixture of dimers and oligomers ranging from
two to nine porphyrin units linked via ether or ester bonds.

Copyright # 2001 John Wiley & Sons, Ltd. J. Porphyrins Phthalocyanines 2001; 5: 161–169
CURRENT STATUS OF PHTHALOCYANINES 163

drawbacks. It is a complex chemical mixture produced by acid ring A) has received market approval in Switzerland
the reaction of hematoporphyrin with 5% sulfuric acid in under the tradename Visudyne1 as a therapy for wet age-
acetic acid followed by treatment with aqueous base and related macular degeneration, a leading cause of blindness
neutralization [27]. The resulting dimers and oligomers, in people over the age of 50. Visudyne1 has also recently
linked primarily with ester and ether bonds, may vary with gained acceptance as a treatment for AMD in several
different preparations and storage times and make struc- countries including the United States and Canada and is
ture–activity relationships impossible to determine [28]. undergoing further clinical trials for other diseases causing
However, since market approval, Photofrin1 is manufac- vision loss due to choroidal neovascularization. Overall, the
tured using stringent chemical composition and stability photosensitizer used to treat a given condition will greatly
guidelines, insuring product homogeneity. It absorbs very depend on the characteristics of the compound and the
weakly at the therapeutic wavelength of 630 nm, where condition to be treated, with a wide range of different
tissue penetration of light is not optimal, limiting treatment photosensitizers being used in clinic, each for its own
to tumor depths of no more than 5 mm [29]. While suitable particular use.
for the more superficial lesions presently being treated,
absorption at longer wavelengths is necessary to treat more
deep-seated or larger tumors. In addition, hematoporphyrin
derivatives have proven to be ineffective for such cancers as PHTHALOCYANINES
pigmented melanoma due to overlapping absorption spectra
of the photosensitizer and melanin in the malignant tissue Among the more promising second-generation photosensi-
[30]. Most importantly, hematoporphyrin derivatives ex- tizers are phthalocyanines (Fig. 2). The term phthalocyanine
hibit an extended retention in cutaneous tissue for up to 10 finds its origin in the Greek word ‘naptha’ which means rock
weeks post-injection [19]. This results in a prolonged skin oil and “cyanine” which means dark blue. It was first used to
photosensitivity, an obvious disadvantage especially for describe this class of macrocyclic compounds by Sir
patients with late-stage malignancies. Nonetheless, the Reginald Linstead in 1933 during his pioneering work on
prolonged skin photosensitivity induced by PDT with the subject [34]. In hindsight, the first recorded observation
Photofrin1 is only a mild inconvenience as compared to of a phthalocyanine occurred in 1907 [35]. During the
the severe adverse effects of standard chemotherapeutic synthesis of o-cyanobenzamide from phthalamide and
regimens. While these disadvantages have not prevented acetic anhydride, Braun and Tcheriac observed the produc-
Photofrin1 from becoming a useful drug against cancer and tion of a colored impurity of unknown structure and origin.
other conditions, the search for new second-generation However, this colored by-product was not further studied. It
photosensitizers with improved physical, chemical and was not until 1927 that a second preparation of phthalo-
spectral properties remains an important goal [23, 31]. cyanine was reported [36]. Scottish Dyes, Ltd. went on to
Ideal photosensitizers for PDT must meet certain criteria, further exploit the usefulness of these macrocycles and was
[9, 27, 29, 32, 33] namely: granted the first patent with respect to phthalocyanines [37].
In addition to the immediate applications as dyes and
. It should be chemically pure and maintain a constant colorants, it was realized that such compounds would be of
composition throughout treatment, undergoing minimal great academic interest. As such, starting in 1929, Linstead
photobleaching. and his group began their work on phthalocyanines which
. It should have minimal dark toxicity. lead to the determination of their structure in the early 1930s
. The photosensitizer should be preferentially retained by [38–40]. The synthesis of Pcs [41–43] and their relationship
the target tissue, whether malignant tumor cells or viral to porphyrins [44], along with further examination into the
components, so as to induce only marginal toxicity to intricate structure of Pcs [43], their planar nature, their
surrounding healthy biological matter. In addition, the complexes with metal ions [40, 43, 45, 46] and their
excess dye should be rapidly excreted from the body stability [38, 43] were subsequently studied. The structure
exhibiting low systemic toxicity. of phthalocyanines was later confirmed by Robertson via
. The dye should have high photochemical reactivity with a
high quantum yield of long-lived triplet-states energetic
enough to produce singlet oxygen.
. The sensitizer should have a large absorption coefficient
at a long wavelength (600–800 nm) where there is optimal
tissue penetration by light with a low degree of
attenuation by hemoglobin. Furthermore, cheaper diode
lasers can be used in this range, thus increasing the
potential utility of PDT in a clinical setting.

While no photosensitizer can be expected to fulfill all of


these parameters, numerous second-generation photosensi-
tizers have been investigated that can overcome the
shortcomings of hematoporphyrin derivatives and take
advantage of their more highly suited properties in order
to treat a number of conditions. For instance, the use of 5-
aminolaevulinic acid (ALA)-induced endogenous photo-
sensitizers has recently gained clinical acceptance for the Fig. 2. Typical phthalocyanine structure where M is the central
treatment of actinic keratoses of the face and scalp. In the metal atom (Al, Co, Ga, Si, Zn) and R represents a multitude of
meanwhile, verteporfin (benzoporphyrin derivative mono- possible ring substituents including SO3H, F, COOH, etc.

Copyright # 2001 John Wiley & Sons, Ltd. J. Porphyrins Phthalocyanines 2001; 5: 161–169
164 C. M. ALLEN ET AL.

X-ray crystallography in a series of classic papers [47–50]. detergents, serum, plasma proteins or organic solvents
In the seventy years to follow this initial work, a plethora of [57, 61].
information emerged concerning diverse synthetic routes,
Pc photochemical and photophysical properties and poten-
tial applications. They have long been used as dyes and Phthalocyanine Delivery
coloring agents [51] and have recently gained an industrial
application as photoconducting agents in photocopying Increased photodynamic action has been attributed to
devices [52]. Other potential applications for phthalo- increased monomerization of the photosensitizer. Lipophilic
cyanines include chemical sensors [53], optical storage photosensitizers require biologically compatible delivery
devices [54], molecular metals and conducting polymers systems. Nanoparticles have received attention as potential
[55] and catalysts for numerous chemical reactions [51]. delivery vehicles for drug targeting. A hexadecafluoro zinc
Of the utmost importance is their role in photodynamic phthalocyanine was formulated in polyethylene–glycol-
therapy. These azaporphyrin derivatives have stronger coated poly(lactic acid) nanoparticles and was evaluated
absorbances at longer wavelengths than do porphyrins. for PDT efficiency. Using the EMT-6 tumor model,
Pcs have favorable photophysical and chemical properties. improved tumor response was observed using the nanopar-
In addition, these properties can be altered through the ticle formulation as compared to the same dye solubilized in
addition of substituents to the periphery of the macrocycle Cremophor EL [62]. Nanoparticle formulation allows for
or axial ligands to the chelated central metal ion. This makes increased serum levels of the dye as compared to
them interesting photosensitizing agents. Cremophor EL formulation. Increased blood levels and
Phthalocyanine derivatives have attractive photophysical enhanced tumor uptake lead to higher tumor-to-organ
and photochemical properties as compared to hematopor- concentration ratios enhancing the overall dye efficacy
phyrin derivatives. Monomeric Pcs have a characteristic [63]. Likewise pH-responsive, polymeric micelles consist-
absorption spectra with a strong Soret absorption peak at ing of random copolymers of N-isopropylacrylamide,
approximately 350 nm, a weak maximum around 600 nm methacrylic acid and octadecyl acrylate have been loaded
and a narrow, very strong, absorption peak in the far-red with water-insoluble AlPcCl and shown to exhibit higher
region of the visible spectra (Pc max  680 nm; HpD photocytotoxicity against EMT-6 cells in vitro than the
max  630 nm) where tissue penetration by visible light is control Cremophor EL preparation [64].
superior. In addition to the red-shift of the phthalocyanine Q In addition to emulsifiers such as Cremophor EL, Solutol
band absoption maxima, Pcs have an improved capacity to HS or lipid-based vehicles such as liposomes, a substantial
absorb light, by two orders of magnitude over that of the amount of research has focused on proteinic carriers for
highest Q band absorption of HpD (Pc molar extinction improved targeting of malignant cells. Serum lipoproteins
coefficient, e  105 Mÿ1 cmÿ1; HpD e  103 Mÿ1 cmÿ1) can incorporate hydrophobic sensitizers into their lipid
[56, 57]. These qualities therefore lead to enhanced photo- portion. Frequently, neoplastic cells have increased low-
physical and photochemical properties. density lipoprotein (LDL) receptors on the cell surface as
The nature of the central metal ion influences the Pc increased proliferation requires cholesterol for membrane
photophysical properties (triplet quantum yield and life- synthesis [65]. Subsequently, LDL are known to deliver
time). Complexation of Pcs with open shell or paramagnetic photosensitizers to neoplastic cells through a receptor
metal ions such as Cu2‡, Co2‡, Fe2‡, Ni2‡, VO2‡, Cr3‡ and internalization mechanism [66–69]. Jori and associates
Pd2‡ give dyes with shortened triplet lifetimes (nanosecond have done extensive studies using ZnPc incorporated into
range) due to increased intersystem crossing back to the liposomes of varying composition. In addition, they have
ground state, which renders the dye photoinactive [58]. Pcs investigated the role of serum lipoproteins and found them
containing closed d shell or diamagnetic metal ions, such as to be effective delivery vehicles for photosensitizers such as
Zn2‡, Al3‡ and Ga3‡, are dyes producing high triplet yields phthalocyanines [70–73].
(fT > 0.4) with long lifetimes (tT > 200 ms) [59]. These Recently, a series of publications has reported the use of a
triplets vary in energy from 110–126 kJ/ molÿ1 which is modified aluminum tetrasulfophthalocyanine covalently
ample energy to generate singlet oxygen (94.5 kJ/ molÿ1) coupled to various proteins via one or two caproic acid
with high quantum yields (f) of approximately 0.3–0.5 [60]. spacer chains. Brasseur et al. targeted the scavenger
Singlet oxygen yield is affected by ring substituents and receptor of macrophages using a maleylated bovine serum
axial ligands. In aqueous solutions, Pcs, either non- albumin (BSA) covalently labeled with AlPcS4. Relative
substituted or substituted with hydrophilic groups, tend to photocytotoxicities were reported using a receptor positive
aggregate or dimerize which affects cell penetrating and a receptor negative cell line, where their lethal effects
properties and results in a loss of photochemical activity. correlated with receptor affinity for the BSA–Pc conjugate
Upon excitation with light, energy is dissipated via internal [74].
conversion to the ground state rather than triplet formation Using the same phthlocyanine derivative, it was found
with subsequent singlet oxygen production [59]. Decreasing that covalently labeling a monoclonal antibody for the
the number of sulfonate substituents increases the potential CD3‡ antigen in a similar manner did not hinder receptor
to aggregate, however monomeric Pc molecules with recognition. There was increased uptake of the antibody–Pc
identical central metal ions, and varying degrees of conjugate in a receptor positive cell line as compared to the
sulfonation, retain the same photochemical activity. The free phthalocyanine and antibody–Pc uptake was less in a
extent of aggregation determines the MPcSn activity [60]. cell line void of receptors [75]. However, the Pc–antibody
Aggregation is easily detected spectroscopically as absorp- conjugates were photoinactive, most likely due to inefficient
tion occurs at shorter wavelengths in the Soret region and subcellular targeting. Employing similar compounds,
there is a 30–50 nm blue-shift of the Q band absorption peak AlPcS4 was covalently coupled to monoclonal antibodies
which appears more broad and less intense. Disaggregation directed against carcinoembryonic antigen (CEA) [76]. In
to form monomeric dyes can be accomplished by adding vivo studies using nude mice bearing human colon

Copyright # 2001 John Wiley & Sons, Ltd. J. Porphyrins Phthalocyanines 2001; 5: 161–169
CURRENT STATUS OF PHTHALOCYANINES 165

carcinoma xenografts (T380) demonstrated increased up- blood components [23, 79]. Axial ligated Pcs have an
take of the antibody–Pc conjugate as compared to the free important advantage over Pcs substituted on the periphery.
Pc. In vitro results were also encouraging using this anti- Not only does the axial ligand impart greater solubility and
body complex. prevent aggregation, additionally these compounds do not
Using the same principle, adenoviral proteins were exist as isomers. As such, obtaining a pure sample is more
employed to target lung cancer cells rich in the appropriate apparent.
class of integrin receptors [77]. It was concluded that Since the first reports in 1993, Pc4 has been studied
adenoviral capsid proteins, principally the penton base, extensively using various cell lines and protocols. Initial
when covalently bound to a modified AlPcS4 are able to reports using V-79 Chinese hamster lung fibroblast indi-
target receptor-bearing cells. However, in vitro photocyto- cated a three-fold increase in Pc4 cytotoxicity as compared
toxicity was limited. Using the EMT-6 murine model, in to a reference AlPc [80]. No difference in intracellular
vivo results were more encouraging using the Pc–adenoviral uptake was observed between these two photosensitizers,
protein conjugates. seeming to indicate that this increased photocytotoxicity
Urizzi and co-workers described parallel studies using was due to differences in the intracellular distribution and/or
both covalently labeled and a non-covalently labeled LDL the aggregation of the phthalocyanines in vitro [81]. Pc4
to target human adenocarcinoma lung cancer cells (A549). was also shown to be highly phototoxic on human erythro-
Covalently labeling the apoprotein portion did not hinder cyte ghosts, photoinducing lipid peroxidation. Using singlet
receptor recognition yet cytotoxic effects were marginal, oxygen quenchers such as sodium azide and histidine, it was
presumably due to photosensitizer entrapment in the determined that Pc4, at least in part, induces cytotoxicity via
endosome. Amphiphilic AlPcS4 molecules bearing long a Type II mechanism [82].
aliphatic side chains on the Pc macrocycle were non- Early in vivo reports were also promising using RIF-1
covalently inserted in the LDL phospholipid layers. There fibrosarcoma tumors in mice as a test model. At reasonable
was a substantial increase in cell uptake of the phthalocya- Pc4 doses (1 mg kgÿ1) in 25% Cremophor EL, tumor
nine resulting in a 10-fold increase in cytotoxicity of the Pc– regression was evident with no re-growth using a light dose
LDL conjugate in vitro [66]. of 135 J cmÿ2 [80, 83, 84]. Cutaneous photosensitivity was
A novel approach to photosensitizer targeting has transient using this photosensitizer, lasting only 24 hours
recently been reported using epidermal growth factor after onset occurring 10 days post-Pc4 administration.
(EGF) [78]. Both AlPc and CoPc were covalently labeled Photosensitivity was no longer a factor. Photofrin1, under
with EGF. Lutsenko and associates report a five-fold similar conditions, invoked skin photosensitivity over a
increase in photocytotoxicity using the EGF–Pc complex period of 4 days. One month post-injection, mice remained
as compared to the uncoupled Pc. Cytotoxicity was sensitive to solar light [85, 86]. Similarly, skin photosensi-
measured in both the human MCF-7 and the murine B16 tivity using a silicon napthalocyanine having two long alkyl
cell lines. One month post-PDT, in vivo studies using the chain axial ligands was investigated. At minimal photo-
melanoma B16 tumors implanted in C57B1/6 mice resulted therapeutic doses of 0.1 mmol kgÿ1, the dye elicited only a
in tumors being half the volume using the CoPc–EGF mild and transient skin photosensitivity which subsided in a
conjugate as compared to untreated tumors or tumors treated few days. Photofrin1, on the other hand, induced a severe
with CoPc alone. reaction, requiring 3 to 8 weeks for complete recovery [87].
This would seem to indicate that such phthalocyanines do
not exhibit an appreciable and prolonged cutaneous tissue
Pc4 uptake.
The photocytotoxicity of Pc4 was also determined against
A promising phthalocyanine receiving a great deal of L5178Y-R leukemic lymphoblasts [88]. Similar results
attention is the silicon Pc, denoted as Pc4, bearing a were obtained as for RIF-1 cells. In addition, this study
long-chain amino axial ligand (HOSiPcOSi(CH3)2 concluded that the number of axial ligands did not appear to
(CH2)3N(CH3)2) (Fig. 3). This silicon phthalocyanine has affect the photocytotoxicity of the compound. Contrarily,
shown promising results both in vitro and in vivo and is the length of the axial ligand was shown to influence
presently entering clinical trials for the treatment of phototoxicity as extended amino axial substituents were less
neoplasms and has been examined for the sterilization of effective against these murine cell lines. It was theorized
that the shorter aminosiloxy ligands in Pc4 lead to binding at
more important intracellular targets.

Apoptosis or Necrosis
Preliminary results were promising using phthalocyanine
derivatives, thus prompting more in-depth studies regarding
mode of cell death. Apoptosis is a notable mode of cell
death in response to PDT using phthalocyanines [89]. Pc4
has been shown to initiate an apoptotic response in several
cell lines including L5178Y-R cells. DNA fragmentation, a
hallmark of apoptosis, was observed [88]. This same
research team demonstrated the importance of cell mem-
brane damage with respect to apoptosis induction. Photo-
damaging the cell membrane initiates a cascade of events
Fig. 3. Silicon phthalocyanine designated as Pc4. such as phospholipase C activation, release of second

Copyright # 2001 John Wiley & Sons, Ltd. J. Porphyrins Phthalocyanines 2001; 5: 161–169
166 C. M. ALLEN ET AL.

messengers and an increase in intracellular calcium con- human lymphoblasts accumulated ceramide and underwent
centration, all leading to cell suicide [90]. Furthermore, an apoptosis after Pc4-PDT. In contrast, Niemann–Pick disease
increase in intracellular calcium concentration also leads to lymphoblasts, which are deficient in acid sphingomyelinase,
the formation of nitric oxide (NO), an important bioactive fail to respond to Pc4-PDT via ceramide accumulation and
signaling molecule. It has been demonstrated that NO is apoptosis [103]. However, addition of exogenous bacterial
involved in PDT-mediated apoptosis [91]. In addition to SMase during Pc4-PDT induced a significant apoptotic
NO, other well-documented apoptotic pathways are in- response. This lead to the hypothesis that SMase may be an
itiated by Pc4-PDT. Cytochrome c is released into the important proapoptotic factor determining responsiveness
cytosol and subsequent activation of proteases [92], now of cells to Pc4-PDT. Previous work showed that Pc4-PDT
known as caspases, resulting in the cleavage of various was able to induce apoptosis in numerous cell lines. Three
proteins including poly(ADP-ribose)polymerase have been cell lines were studied in vitro revealing the accumulation of
documented [93]. Cell cycle arrest in the G0/G1 phase of the ceramide post-PDT and induction of apoptosis. Interest-
cell cycle as a result of p21 induction has been observed ingly, there were increased levels of ceramide in RIF-1 cells
following Pc4 therapy [94]. In addition, the participation of post-PDT with no evidence of apoptosis. Ceramide clearly
retinoblastoma protein has been studied following photo- is associated with PDT-mediated cell death either by
therapy [95]. Several other apoptotic pathways and proteins inducing apoptosis or necrosis [104].
involved in PDT-mediated apoptosis are under investiga-
tion.
Furthermore, enhanced apoptotic response to photody- Indirect or Direct Effect
namic therapy with AlPc was observed after Bcl-2
transfection [96]. Bcl-2 is a known inhibitor of apoptosis Aluminum disulfonated phthalocyanine with sulfonate
via antagonizing the release of mitochrondrial cytochrome groups on adjacent isoindoline units (AlPcS2adj) has been
c. In this case, using immortalized MCF-10A cells, over- used to photoinactivate human epidermal keratinocyte (UP)
expression of Bcl-2 and bax was revealed and PDT caused and oral squamous cell carcinoma-derived cells (H376) in
selective destruction of Bcl-2, leaving bax unaffected. As vitro. This direct cell kill in vitro suggests that direct killing
such, the greater apoptotic response towards PDT in this of tumor cells may contribute to tumor regression in vivo
case is probably attributed to the higher bax:Bcl-2 ratio after [105]. Using the murine EMT-6 model, disulfonated
photodamage. On the other hand, CHO-K1 cells transfected aluminum phthalocyanine induced a significant direct tumor
with Bcl-2 exhibited a partial resistance to PDT-induced cell phototoxic effect. Tumor vasculature damage, an
cell death, whether via apoptosis or non-apoptotic mechan- indirect effect, was assessed using 99mTc–methoxyisobutyl
isms [97]. isonitrile (99mTc–MIBI) retention in the tumor. The blood
Subsequently, it was demonstrated that mode of cell flow declined to 50% of that of the control tumor and
death is not only dependant on cell type (i.e. fibroblasts do remained constant. This is indicative of direct cell kill as
not die via organized DNA fragmentation [88]) but also on opposed to tumor vasculature shut down. Photofrin1-
the photosensitizer, its intracellular localization and the treated tumors immediately exhibited decreased blood flow
drug and energy dose administered. In addition, it has to the treated tumor with almost no blood entering the tumor
recently be shown that the mode of cell death also varies stroma 24 hours post-PDT. This is indicative of an indirect
with cell density due to the bystander effect [98]. Using tumor cell death by starving the cells of vital nutrients
AlPc and lower PDT doses resulted in the induction of [106, 107]. A recent study confirmed these results using
apoptosis as opposed to necrosis. Organelles such as the positron emission tomography (PET). Mice-bearing EMT-6
mitochondria and lysosomes were mainly affected. At tumors were subjected to either Pc-PDT or PII-PDT. Thirty
higher PDT doses, cells underwent severe membrane minutes post-Pc-PDT, [18F]fluoro-2-deoxy-D-glucose
damage resulting in necrosis [99]. Interestingly, Dellinger (FDG) activity falls to about 50% of the control tumor
reported a mixed response when using Photofrin1 and CV-1 value while remaining well above the blood activity level.
cells [100]. At higher irradiation doses, the cells lysed In contrast, at 2 h post-irradiation, tumor activity drops to
instead of exhibiting fragmentation associated with apop- the blood activity level. These results are consistent with an
tosis. However, morphological changes typical of apoptosis action mechanism proceeding primarily by initial direct cell
were observed initially, such as plasma membrane blebbing, kill, resulting in a gradual decrease in tumor cell survival
condensation of the cytoplasm and nuclear fragmentation. while sparing tumor microvasculature. PII-PDT affected
These cells appear to enter an apoptosis-like death, yet this tumor metabolism immediately following PDT, as seen by
pathway is stopped by plasma membrane lysis and limited FDG uptake, thus suggesting reduced blood flow
ultimately, the cells undergo necrosis. In vivo studies using and poor delivery of the tracer at the tumor site, indicative of
the murine MS-2 fibrosarcoma tumor model demonstrated an indirect cell death mechanism [108].
random necrosis of tumor cells post-ZnPc-PDT. Interest-
ingly, apoptotic pathways were also detected, suggesting
different localization of photosensitizers within the cell Phthalocyanines In-Clinic
upon illumination [101].
Photodynamic therapy using phthalocyanines is known to As was stated previously, Pc4 is about to enter clinical trials
induce a number of responses that ultimately lead to for the treatment of neoplasms and has been investigated for
apoptosis. For instance, PDT using Pc4 induces an oxidative the sterilization of blood components [23, 78]. In addition, a
stress associated with increased ceramide generation [102], liposomal preparation of zinc phthalocyanine (CGP55847)
which has been associated with apoptosis in several malig- has been in early Phase I/II clinical trials in Switzerland for
nant and non-malignant cell lines. The increased production the treatment of patients with squamous cell carcinomas of
of ceramide is due to the stress-induced activation of the upper digestive tract [109]. Attempts were also under-
sphingomyelinase (SMase). It has been shown that normal way to develop a topical formulation of this photosensitizer

Copyright # 2001 John Wiley & Sons, Ltd. J. Porphyrins Phthalocyanines 2001; 5: 161–169
CURRENT STATUS OF PHTHALOCYANINES 167

for use in the treatment of skin disorders such as psoriasis 7. Pfeiffer H. In Handbuch der Biochemischen Arbeits-
[110]. mithoden, vol. 1. Abderhalden E. (ed). Urban &
The onocological center of the Russian Academy of Schwarzenberg: Berlin, 1911; 563.
Medical Sciences and the Moscow Medical Academy 8. Meyer-Betz F. Duetshces. Arch. Klin. Med. 1913; 112:
treated 42 patients from 1992 to 1995 using a mixture of 476.
AlPcSn, trade named Photosense. Cancers such as basal and 9. Bonnett R. Rev. Contemp. Pharmacother. 1999; 10: 1.
squamous cell skin cancers, breast cancer, cancer of the 10. Oschner M. J. Photochem. Photobiol. B: Biol. 1997; 39: 1.
tongue, oral mucous and lower lip, lung and larynx cancer 11. Skalkos D, Hampton JA, Keck RW, Wagoner M, Selman
and various others have been treated using dye doses of SH. J. Photochem. Photobiol. 1994; 59: 175.
0.5–1 mg kgÿ1 body weight with success. Fifty percent 12. Sharman WM, Allen CM, van Lier JE. Methods Enzymol.
underwent complete tumor regression without re-growth 2000; 319: 376.
while 43% exhibited a partial response. Only 7% gave no 13. Weishaupt KR, Gomer CJ, Dougherty TJ. Cancer Res.
response at all [25]. The phthalocyanine mixture exhibited 1976; 36: 2326.
no systemic toxicity. No immune system modifications were 14. Valenzo DP. Photochem. Photobiol. 1987; 46: 147.
observed [111]. More precisely, 93 tumor sites with 15. Fuchs J, Thiele J. Free Radic. Biol. Med. 1998; 24: 835.
different head and neck tumors were treated with Photo- 16. Moan J, Boye E. Photobiochem. Photobiophys. 1981; 2:
sense. Over 50% of the tumors exhibited a complete 301.
response where there was no clinical evidence of malignant 17. Moan J. J. Photochem. Photobiol. B: Biol. 1990; 6: 343.
tissue. Various cancer types responded well to Photosense 18. Rosenthal I, Ben Hur E. Int. J. Radiat. Biol. 1995; 67: 85.
therapy except for melanoma, where none of the treated 19. Philips D. Prog. Reaction Kinetics 1997; 22: 175.
neoplasms gave a complete response [112]. The State 20. Gantchev TG, van Lier JE. Photochem. Photobiol. 1995;
Research Center for Laser Medicine (Moscow, Russia) 62: 123.
reported excellent results for skin cancer when using the Pc- 21. Hadjur C, Wagnières G, Ihringer F, Monnier Ph, van den
PDT. Overall, 85% responded completely to the Pc-PDT Bergh H. J. Photochem. Photobiol. B: Biol. 1997; 38: 196.
treatment of skin cancers [24]. A recent review of phthalo- 22. Viola A, Jeunet A, Decreau R, Chanon M, Julliard M.
cyanines as photosensitizers has been published outlining Free Radic. Res. 1998; 28: 517.
the success of the clinical studies carried out in Russia 23. Sharman WM, Allen CM, van Lier JE. Drug Discovery
[113]. Today 1999; 4: 507.
24. Sobolev AS, Stranadko EF. Int. Photodynamics 1997; 1:
2.
CONCLUSION 25. Stranadko EF, Skobelkin OK, Litvin GD, Astrakhankina
TA. SPIE 1996; 2728: 194.
In this new millennium, photodynamic therapy is becoming 26. Sokolov VV, Chissov VI, Filonenko EV, Sukhin GM,
a tangible treatment not only for malignancies but for Yakubovskaya RI, Belous TA, Zharkova NN, Kozlov DN,
various afflictions too numerous to mention in this Smirov VV. SPIE 1994; 2325: 367.
publication [23, 114–122]. It is interesting that in this age 27. Bonnett R. Chem. Soc. Rev. 1995; 24: 19.
of technology the simplicity of this bimodal treatment 28. Brown SB, Truscott TG. Chem. Br. 1993; 29: 955.
contributes to its versatility and usefulness. Phthalocyanines 29. Wöhrle D, Hirth A, Bogdahn-Rai T, Schnurpfeil G,
are extremely powerful photosensitizers and pre-clinical Shopova M. Russ. Chem. Bull. 1998; 47: 807.
work clearly show their immense promise in photodynamic 30. Peeva M, Shopova M, Stoichkova N, Michailov N,
therapy. Only the future will dictate whether phthalo- Wöhrle D, Müller S. J. Porprhyrins Phthalocyanines
cyanines will fulfill their potential as photosensitizing 1999; 3: 380.
agents for the treatment of cancer and other conditions. 31. Ali H, van Lier JE. Chemical Rev. 1999; 99: 2379.
32. van Lier JE. In Light in Biology and Medicine. Douglas
Acknowledgements RH, Moan J, Dall’ Acqua F. (eds). Plenum: New York,
1988; 133.
The Canadian Institutes of Health Research is gratefully 33. MacRoberts AJ, Bown SG, Philips D. In Photosensitizing
acknowledged for their financial support. J.E.vL. is the Compounds: Their Chemistry, Biology and Clinical Use.
holder of the Jeanne and J.-Louis Lévesque Chair in Bock G, Harnett S. (eds). Wiley: Chicester, 1989; 4.
Radiobiology. C.M.A. and W.M.S. are recipients of 34. Linstead RP. J. Chem. Soc. 1934; 1016.
doctoral stipends from the Minister of Education of Québec 35. Braun A, Tscherniac J. Ber. 1907; 40: 2709.
(Fonds pour la formation de Chercheurs et l’Aide á la 36. de Diesbach H, von der Weid E. Helv. Chim. Acta 1927;
Recherche, FCAR). 10: 886.
37. Dandridge AG, Drescher HA, Thomas J. British Patent
322, 169, (Scottish Dyes, Ltd.) May 16, 1929.
REFERENCES 38. Linstead RP, Lowe AR. J. Chem. Soc. 1934; 1031.
39. Dent CE, Linstead RP, Lowe AR. J. Chem. Soc. 1934;
1. Daniell MD, Hill JS. Aust. N. Z. J. Surg. 1991; 61: 340. 1033.
2. Raab O. Z. Biol. 1900; 39: 524. 40. Linstead RP, Robertson JM. J. Chem. Soc. 1936; 1736.
3. von Tappeiner H, Jodlbauer A. Vogel FGW, 1907; 41. Byrne GT, Linstead RP, Lowe AR. J. Chem. Soc. 1934;
Leipzig, p. 210. 1017.
4. Hausmann W. Wien. Klin. Wchschr. 1908; 21: 1527. 42. Linstead RP, Lowe AR. J. Chem. Soc. 1934; 1022.
5. Hausmann W. Wien. Klin. Wchschr. 1908; 22: 1820. 43. Dent CE, Linstead RP. J. Chem. Soc. 1934; 1027.
6. Hausmann W. Biochem. Ztschr. 1911; 30: 276. 44. Dent CE. J. Chem. Soc. 1938; 1.

Copyright # 2001 John Wiley & Sons, Ltd. J. Porphyrins Phthalocyanines 2001; 5: 161–169
168 C. M. ALLEN ET AL.

45. Barrett PA, Dent CE, Linstead RP. J. Chem. Soc. 1936; 77. Allen CM, Sharman WM, La Madeleine C, Weber JM,
1719. Langlois R, Ouellet R, van Lier JE. Photochem. Photo-
46. Barrett PA, Frye DA, Linstead RP. J. Chem. Soc. 1938; biol. 1999; 70: 512.
1157. 78. Lutsenko SV, Feldman NB, Finakova GV, Posypanova
47. Robertson JM. J. Chem. Soc. 1935; 615. GA, Severin SE, Skryabin KG, Kirpichnikov MP,
48. Robertson JM. J. Chem. Soc. 1936; 1195. Lukyanets EA, Vorozhtsov GN. Tumor Biol. 1999; 20:
49. Robertson JM, Woodard I. J. Chem. Soc. 1937; 219. 218.
50. Robertson JM, Woodard I. J. Chem. Soc. 1940; 36. 79. Egorin MJ, Zuhowski EG, Sentz DL, Dobson JM, Callery
51. Moser FH, Thomas AL. In The Phthalocyanines, vol. 1 PS, Eiseman JL. Cancer Chemother. Pharmacol. 1999;
and 2. CRC: Boca Raton, 1983. 44: 283.
52. Leznoff CC, Lever ABP. In Phthalocyanines: Properties 80. Oleinick NL, Antunez AR, Clay ME, Rihter BD, Kenney
and Applications, vol. 4. Leznoff CC, Lever ABP. (eds). ME. Photochem. Photobiol. 1993; 57: 242.
VCH: New York, 1996 p. v. 81. He J, Horng M-F, Deahl JT, Oleinick NL, Evans HH.
53. Snow AW, Barger WR. In Phthalocyanines: Properties Photochem. Photobiol. 1998; 67: 720.
and Applications, vol. 1. Leznoff CC, Lever ABP. (eds). 82. Zaidi SIA, Agarwal R, Eichler G, Rihter BD, Kenney ME,
VCH: New York, 1989; 341. Mukhtar H. Photochem. Photobiol. 1993; 58: 204.
54. Shirk JS. In Phthalocyanines: Properties and Applica- 83. Zaidi SIA, Oleinick NL, Zaim MT, Mukhtar H. Photo-
tions, vol. 1. Leznoff CC, Lever ABP. (eds). VCH: New chem. Photobiol. 1993; 58: 771.
York, 1989; 79. 84. Agarwal R, Korman NJ, Mohan RR, Feyes DK, Jawed S,
55. Wöhrle D. In Phthalocyanines: Properties and Applica- Zaim MT, Mukhtar H. Photochem. Photobiol. 1996; 63:
tions, Vol. 1. Leznoff CC, Lever ABP. (eds). VCH: New 547.
York, 1989; 55. 85. Anderson C, Hrakbovsky S, McKinley Y, Tubesing K,
56. van Lier JE, Brasseur N, Paquette B, Wagner JR, Ali H, Tang H-P, Dunbar R, Mukhtar H, Elmets CA. Photochem.
Langlois R, Rousseau J. In Photosensitization, Moreno G. Photobiol. 1997; 65: 895.
(ed). Springer-Verlag: Berlin, 1988; 435. 86. Anderson CY, Freye K, Tubesing KA, Li Y-S, Kenney
57. van Lier JE, Spikes J. In Photosensitizing Compounds: ME, Mukhtar H, Elmets CA. Photochem. Photobiol.
Their Chemistry, Biology and Clinical Use, Wiley: 1998; 67: 332.
Chichester, 1989; 95. 87. Brasseur N, Ouellet R, Lewis K, Potter WR, van Lier JE.
58. Chan WS, Marshall JF, Svensen R, Phillips D, Hart IR.
Photochem. Photobiol. 1995; 62: 1058.
Photochem. Photobiol. 1987; 45: 757.
88. He J, Larkin HE, Li Y-S, Rihter BD, Zaidi SIA, Rodgers
59. Darwent JR, Douglas P, Harriman A, Porter G, Richoux
MAJ, Mukhtar H, Kenney ME, Oleinick NL. Photochem.
M–C. Coord. Chem. Rev. 1982; 44: 83.
Photobiol. 1997; 65: 581.
60. Wagner JR, Ali H, Langlois R, Brasseur N, van Lier JE.
89. Oleinick NL. Photodynamic News 1998; 1: 6.
Photochem. Photobiol. 1987; 45: 587.
90. Agarwal ML, Larkin HE, Zaidi SIA, Mukhtar H, Oleinick
61. Ben Hur E, Siwecki JA, Newman HC, Crane SW,
NL. Cancer Res. 1993; 53: 5897.
Rosenthal I. Cancer Lett. 1987; 38: 215.
91. Gupta S, Ahmad N, Mukhtar H. Cancer Res. 1998; 58:
62. Allémann E, Rousseau J, Brasseur N, Kudrevich SV,
1785.
Lewis K, van Lier JE. Int. J. Cancer 1996; 66: 821.
63. Allémann E, Brasseur N, Benrezzak O, Rousseau J, 92. Varnes ME, Chiu S-M, Xue L-Y, Oleinick NL. Biochem.
Kudrevich SV, Boyle RW, Leroux JC, Gurny R, van Lier Biophys. Res. Commun. 1999; 255: 673.
JE. J. Pharm. Pharmacol. 1995; 47: 382. 93. He J, Whitacre CM, Xue L-Y, Berger NA, Oleinick NL.
64. Taillefer J, Jones M–C, Brasseur N, van Lier JE, Leroux J- Cancer Res. 1998; 58: 940.
C. J. Pharm. Sci. 2000; 89: 62. 94. Ahmad N, Feyes DK, Agarwal R, Mukhtar H. Proc. Natl.
65. Vitols S. Cancer Cells 1991; 3: 488. Acad. Sci. USA 1998; 95: 6977.
66. Urizzi P, Allen CM, Langlois R, Ouellet R, La Madeleine 95. Ahmad N, Gupta S, Mukhtar H. Oncogene 1999; 18:
C, van Lier JE. J. Porphyrins Phthalocyanines 2000; 4: 1. 1891.
67. Hamblin MR, Newman EL. J. Photochem. Photobiol. B: 96. Choi Kim H-R, Luo Y, Li G, Kessel D. Cancer Res. 1999;
Biol. 1994; 26: 147. 59: 3429.
68. Mazière JC, Molière P, Santos R. J. Photochem. 97. He J, Agarwal ML, Larkin HE, Friedman LR, Xue L-Y,
Photobiol. B: Biol. 1991; 8: 351. Oleinick NL. Photochem. Photobiol. 1996; 64: 845.
69. Allison BA, Pritchard PH, Levy JG. Br. J. Cancer 1994; 98. Dahle J, Steen HB, Moan J. Photochem. Photobiol. 1999;
69: 833. 70: 363.
70. Polo L, Bianco G, Reddi E, Jori G. Int. J. Biochem. Cell 99. Luo Y, Kessel D. Photochem. Photobiol. 1997; 66: 479.
Biol. 1995; 27: 1249. 100. Dellinger M. Photochem. Photobiol. 1996; 64: 182.
71. Reddi E. J. Photochem. Photobiol. B: Biol. 1997; 37: 189. 101. Jori G, Fabris C. J. Photochem. Photobiol. B: Biol. 1998;
72. Reddi E, Zhou C, Biolo R, Menegaldo E, Jori G. Br. J. 43: 181.
Cancer 1990; 61: 407. 102. Separovic D, He J, Oleinick NL. Cancer Res. 1997; 57:
73. Valduga G, Pifferi A, Taroni P, Valentini G, Reddi E, 1717.
Cubeddu R, Jori G. J. Photochem. Photobiol. B: Biol. 103. Separovic D, Pink JJ, Oleinick NA, Kester M, Boothman
1999; 49: 198. DA, McLoughlin M, Pena LA, Haimovitz-Friedman A.
74. Brasseur N, Langlois R, La Madeleine C, Ouellet R, van Biochem. Biophys. Res. Commun. 1999; 258: 506.
Lier JE. Photochem. Photobiol. 1999; 69: 345. 104. Separovic D, Mann KJ, Oleinick NL. Photochem.
75. Ménard I. MSc Thesis, Université de Sherbrooke, 1998. Photobiol. 1998; 68: 101.
76. Carcenac M, Larroque C, Langlois R, van Lier JE, Artus 105. Ketabchi A, MacRoberts A, Speight PM, Bennett JH.
JC, Pélegrin A. Photochem. Photobiol. 1999; 70: 930. Arch. Oral Biol. 1998; 43: 143.

Copyright # 2001 John Wiley & Sons, Ltd. J. Porphyrins Phthalocyanines 2001; 5: 161–169
CURRENT STATUS OF PHTHALOCYANINES 169

106. Chan W-S, Brasseur N, La Madeleine C, van Lier JE. 113. Lukyanets EA. J. Porphyrins Phthalocyanines 1999; 3:
Anticancer Res. 1996; 16: 1887. 424.
107. Chan W-S, Brasseur N, La Madeleine C, Ouellet R, van 114. van Hillegersberg R, Kort WJ, Wilson JHP. Drugs 1994;
Lier JE. Eur. J. Cancer 1997; 33: 1855. 48: 510.
108. Lapointe D, Brasseur N, Cadorette J, La Madeleine C, 115. Dougherty TJ, Gomer CJ, Henderson BW, Jori G, Kessel
Rodrigue S, van Lier JE, Lecomte R. J. Nucl. Med. 1999; D, Korbelik M, Moan J, Peng Q. J. Natl. Can. Inst. 1998;
40: 876. 90: 889.
109. Oschner M. J. Photochem. Photobiol. B: Biol. 1996; 32: 3. 116. Rivellese MJ, Baumal CR. Opthal. Surg. Lasers 1999; 30:
110. Levy JG. Trends Biotechnol. 1995; 13: 14. 653.
111. Sokolov VV, Chissov VI, Filonenko EV, Yakubovskaya 117. Kessel D, Dougherty TJ. Rev. Contemp. Pharmacother.
RI, Sukhin DG, Galpern MG, Vorozhtsov GN, Gulin AV, 1999; 10: 19.
Zhitkova MB, Zharkova NN, Kozlov DN, Smirnov VV. 118. Okunaka T, Kato H. Rev. Contemp. Pharmacother. 1999;
Proc. SPIE 1994; 2325: 364. 10: 59.
112. Vakoulovskaia EG, Chental VV, Abdoullin NA, 119. Hahn SM, Glatstein E. Rev. Contemp. Pharmacother.
Kuvshinov YP, Tabolinovskaia TD, Edinak NJ, Poddubny 1999; 10: 69.
BK, Lioubaev VL, Boikov VP, Kondratjeva TT, 120. McCaughan JS Jr.. Drugs and Aging 1999; 15: 49.
Meerovich GA, Stratonnikov AA, Linkov KG, Agafonov 121. His RA, Rosenthal DI, Glatstein E. Drugs 1999; 57: 725.
VV. SPIE 1996; 2924: 309. 122. Stables GI. J. Dermatol. Treat. 1999; 10: 213.

Copyright # 2001 John Wiley & Sons, Ltd. J. Porphyrins Phthalocyanines 2001; 5: 161–169

You might also like