throbber
OPEN ACCESS
`
`
`
`molecules
`
`Molecules 2011, 16, 4140-4164; doi:10.3390/molecules16054140

`
`ISSN 1420-3049
`www.mdpi.com/journal/molecules
`
`Review
`Aminolevulinic Acid (ALA) as a Prodrug in Photodynamic
`Therapy of Cancer
`
`Małgorzata Wachowska 1, Angelika Muchowicz 1, Małgorzata Firczuk 1,
`Magdalena Gabrysiak 1, Magdalena Winiarska 1, Małgorzata Wańczyk 1, Kamil Bojarczuk 1 and
`Jakub Golab 1,2,*
`
`1 Department of Immunology, Centre of Biostructure Research, Medical University of Warsaw,
`Banacha 1A F Building, 02-097 Warsaw, Poland
`2 Department III, Institute of Physical Chemistry, Polish Academy of Sciences, 01-224 Warsaw,
`Poland
`
`* Author to whom correspondence should be addressed; E-Mail: jakub.golab@wum.edu.pl;
`Tel. +48-22-5992199; Fax: +48-22-5992194.
`
`Received: 3 February 2011 / Accepted: 3 May 2011 / Published: 19 May 2011
`
`
`Abstract: Aminolevulinic acid (ALA) is an endogenous metabolite normally formed in the
`mitochondria from succinyl-CoA and glycine. Conjugation of eight ALA molecules yields
`protoporphyrin IX (PpIX) and finally leads to formation of heme. Conversion of PpIX to
`its downstream substrates requires the activity of a rate-limiting enzyme ferrochelatase.
`When ALA is administered externally the abundantly produced PpIX cannot be quickly
`converted to its final product - heme by ferrochelatase and therefore accumulates within
`cells. Since PpIX is a potent photosensitizer this metabolic pathway can be exploited in
`photodynamic therapy (PDT). This is an already approved therapeutic strategy making
`ALA one of the most successful prodrugs used in cancer treatment.
`
`Key words: 5-aminolevulinic acid; photodynamic therapy; cancer; laser; singlet oxygen
`
`
`
`1. Introduction
`
`
`Photodynamic therapy (PDT) is a minimally invasive therapeutic modality used in the management
`of various cancerous and pre-malignant diseases. It involves the systemic administration of a non-toxic
`photosensitizing (PS) drug, which accumulates in host and tumor cells, and subsequent illumination of
`
`

`

`OPEN ACCESS
`
`
`
`molecules
`
`Molecules 2011, 16, 4140-4164; doi:10.3390/molecules16054140

`
`ISSN 1420-3049
`www.mdpi.com/journal/molecules
`
`Review
`Aminolevulinic Acid (ALA) as a Prodrug in Photodynamic
`Therapy of Cancer
`
`Małgorzata Wachowska 1, Angelika Muchowicz 1, Małgorzata Firczuk 1,
`Magdalena Gabrysiak 1, Magdalena Winiarska 1, Małgorzata Wańczyk 1, Kamil Bojarczuk 1 and
`Jakub Golab 1,2,*
`
`1 Department of Immunology, Centre of Biostructure Research, Medical University of Warsaw,
`Banacha 1A F Building, 02-097 Warsaw, Poland
`2 Department III, Institute of Physical Chemistry, Polish Academy of Sciences, 01-224 Warsaw,
`Poland
`
`* Author to whom correspondence should be addressed; E-Mail: jakub.golab@wum.edu.pl;
`Tel. +48-22-5992199; Fax: +48-22-5992194.
`
`Received: 3 February 2011 / Accepted: 3 May 2011 / Published: 19 May 2011
`
`
`Abstract: Aminolevulinic acid (ALA) is an endogenous metabolite normally formed in the
`mitochondria from succinyl-CoA and glycine. Conjugation of eight ALA molecules yields
`protoporphyrin IX (PpIX) and finally leads to formation of heme. Conversion of PpIX to
`its downstream substrates requires the activity of a rate-limiting enzyme ferrochelatase.
`When ALA is administered externally the abundantly produced PpIX cannot be quickly
`converted to its final product - heme by ferrochelatase and therefore accumulates within
`cells. Since PpIX is a potent photosensitizer this metabolic pathway can be exploited in
`photodynamic therapy (PDT). This is an already approved therapeutic strategy making
`ALA one of the most successful prodrugs used in cancer treatment.
`
`Key words: 5-aminolevulinic acid; photodynamic therapy; cancer; laser; singlet oxygen
`
`
`
`1. Introduction
`
`
`Photodynamic therapy (PDT) is a minimally invasive therapeutic modality used in the management
`of various cancerous and pre-malignant diseases. It involves the systemic administration of a non-toxic
`photosensitizing (PS) drug, which accumulates in host and tumor cells, and subsequent illumination of
`
`

`

`Molecules 2011, 16
`
`
`
`4141
`
`the tumor site with visible light, corresponding to the appropriate photosensitizer absorption
`wavelength (Figure 1).
`
`Figure 1. Overview of PDT. Following photosensitizer administration it undergoes
`systemic distribution and selectively accumulates in the tumor. Illumination activates the
`photosensitizer and in the presence of molecular oxygen triggers a photochemical reaction
`that culminates in the production of 1O2.
`
`
`
`
`The excited photosensitizer contributes to the generation of singlet oxygen and other reactive
`oxygen species (Figure 2), which results in the oxidative damage to intracellular macromolecules and
`consequently leads to cell death. The mode of PDT-induced cell death is usually a mixture of apoptosis,
`necrosis and autophagy, with the dominance of a particular process depending on the PS (mainly its
`subcellular localization) as well as light fluence. It is generally agreed that apart from the direct
`cellular cytotoxicity, two other important factors contribute to the overall PDT effect: the vascular
`shutdown and local inflammatory reaction [1-4]. One of the major advantages of PDT over other
`anticancer treatment modalities is its high degree of selectivity. This is accomplished via the
`combination of two inactive components, visible light and a photosensitizing drug, which applied
`together in the presence of oxygen lead to generation of cytotoxic intermediates that effectively kill
`tumor cells [5].
`
`
`
`

`

`Molecules 2011, 16
`
`
`
`4142
`
`Figure 2. Types of oxidative reactions during PDT. Light with sufficient energy and
`wavelength matching absorption spectrum of the photosensitizer (PS) can activate a
`photochemical reaction that leads to formation of activated PS molecule (denoted by
`asterisk). Activated PS can lose its energy by emitting of visible light (fosforescence or
`fluorescence). Alternatively it may generate singlet oxygen in a type II reaction or free
`radicals in a type I reaction.
`
`As the PS alone is non-toxic and ineffective, to some extent it can be considered as a prodrug.
`However, additional selectivity of PDT may be achieved by the administration of a PS precursor. The
`only clinically approved example of such a compound is δ-aminolevulinic acid (ALA), a precursor of
`the natural photosensitizer phrotoporphyrin IX (PpIX). In contrast to exogenously administered PSs
`such as Photofrin, the photodynamically inactive, non-selective and non-toxic compound, ALA, is
`intracellularly metabolized to the photodynamically active PpIX. Subsequent illumination of the tumor
`site with red light activates PpIX, triggers the oxidative damage and induces cytotoxicity [6].
`
`
`
`

`

`Molecules 2011, 16
`
`
`
`4143
`
`ALA is a naturally occurring compound, the early intermediate in the heme biosynthesis pathway.
`For therapeutic purposes ALA is administered topically or systemically and penetrates non-selectively
`into all cells, where it is metabolized to an active sensitizer PpIX. The bioactivation of ALA utilizes
`the enzyme machinery of the heme biosynthesis pathway. Although nearly all human cell types
`express the enzymes involved in the heme synthesis, a distinct activity of the enzymes in tumor as
`compared with normal cells leads to a higher PpIX accumulation within transformed cells [7].
`For the last two decades a substantial amount of research has been focused on the elucidation of the
`mechanism of ALA-PDT and the improvement of its therapeutic activity. ALA is a polar molecule and
`in physiological pH occurs mainly as a charged zwitterion, which accounts for its low lipid solubility
`and reduced bioavailability. Further modifications of ALA aimed at improving its cellular permeability,
`increased stability in physiologic pH, increased selectivity and limitation of side effects, are important
`challenges in order to extend the clinical use of ALA-PDT. Since the very first topical application of
`ALA in the treatment of basal cell carcinoma in 1990 [8], the clinical use of ALA-PDT is still growing.
`Nowadays, PDT with ALA and its esters is an approved treatment of several malignant and
`premalignant conditions such as actinic keratosis, basal cell carcinoma, Bowen’s disease, bladder
`cancer and others. This review presents the use of ALA and its derivatives as prodrugs in PDT and
`summarizes the preclinical and clinical results of the treatment.
`
`2. Metabolism of ALA
`
`2.1. Heme Biosynthesis
`
`The synthesis of ALA is the first and rate-limiting step in the biosynthesis of heme [9-11]. ALA is
`normally synthesized in mitochondria in the condensation reaction between glycine and succinyl-CoA
`(Figure 3). The reaction is catalyzed by ALA synthase (ALAS) and requires pyridoxal-5-phosphate
`(PLP) as a cofactor. In mammals, two isoforms of ALA synthase have been identified: ALAS1, which
`is a housekeeping enzyme and ALAS2, which is expressed only in erythroid precursors [12].
`After being synthesized ALA reaches cytosol, where it undergoes a condensation reaction. The
`
`reaction occurs between two ALA molecules with the aid of zinc-dependent enzyme – aminolevulinate
`
`dehydratase (ALAD) – and leads to the formation of porphobilinogen (PBG). ALAD, also known as
`porphobilinogen synthase (PBGS) comprises four homodimers, each of them having one active
`site [13]. Two molecules of ALA bind non-symmetrically to the active site, finally leading to the
`synthesis of PBG [14].
`The next step in heme biosynthesis involves combining four molecules of PBG to form an unstable
`tetrapyrolle - hydroxymethylbilane (HMB). The reaction is catalyzed by porphobilinogen deaminase
`(PBDG), an enzyme containing dipyrromethane in its active site. Dipyrromethane is a co-factor
`covalently bound to the enzyme and consists of two PBG molecules. Four additional molecules of
`PBG attach to dipyrromethane leading to the formation of hexapyrolle. Afterwards, in the hydrolytic
`reaction, cleavage of the distal tetrapyrolle occurs, resulting in the release of HMB [15].
`Hydroxymethylbilane can then enter two pathways. The first one uses uroporphyrinogen III synthase
`(URO3S) to close the HMB macrocycle leading to conversion of tetrapyrolle to uroporphyrinogen III.
`Alternatively, HMB can undergo spontaneous cyclization, which leads to the formation of
`uroporphyrinogen I [16].
`
`

`

`Molecules 2011, 16
`
`
`
`4144
`
`Figure 3. Heme biosynthesis. The graphic presents the most important steps in the heme
`biosynthetic pathway.
`
`
`
`
`Uroporphyrinogen decarboxylase (UROD) catalyzes decarboxylation of all four acetate side chains
`of uroporphyrinogen III to methyl groups [17]. The product of this reaction – coproporphyrinogen III
`is transported to the intermembrane space of mitochondria probably by peripheral-type benzodiazepine
`receptors (PBR) [18,19]. Coproporphyrinogen oxidase (CPO) then catalyzes the conversion of
`coporoporphyrinogen III to protoporporphyrinogen IX with the release of H2O2 and CO2. The reaction
`provides vinyl groups by oxidative decarboxylation of propionate groups on pyrolle rings A and
`B [20]. There are two forms of CPO: oxygen-dependent found in aerobic organisms, and
`oxygen-independent, in anaerobic and facultative bacteria [21]. In mammals, oxygen-dependent CPO
`is originally synthesized in cytosol with unusually long N-terminal sequence targeting it to the
`mitochondria [22]. Finally, CPO is situated in the mitochondrial intermembrane space with a fraction
`loosely bound to the inner membrane [23].
`The next intermediate in heme biosynthesis, protoporphyrin IX (PpIX), is synthesized in the
`mitochondria and requires FAD-containing protoporphyrinogen oxidase (PPO) [24,25]. PPO catalyzes
`conversion of protoporphyrinogen IX to PpIX in the six-electron oxidation. The reaction requires
`oxygen as a terminal electron acceptor and leads to removal of six hydrogens from protoporphyrinogen
`IX [7]. Ferrochelatase (FECH), another rate-limiting enzyme, is responsible for insertion of Fe2+ into
`PpIX. The reaction occurs on the inner surface of the inner mitochondrial membrane [26]. This stage
`leads to the formation of the final product – heme – and completes the heme biosynthetic pathway.
`
`

`

`Molecules 2011, 16
`
`2.2. Heme Degradation
`
`
`
`4145
`
`The enzyme responsible for heme degradation is heme oxygenase (HO). HO exists in two izoforms,
`significantly different in their regulatory mechanism: HO-1 is an inducible form whereas HO-2 is
`expressed constitutively [27,28]. They both catalyze the same reaction, which leads to the production
`of biliverdin, carbon monoxide (CO) and iron [29,30]. The reaction involves formation of HO-heme
`complex and reduction of ferric heme-iron to Fe2+ by NADPH:cytochrome p-450 reductase [31].
`Afterwards, three oxygenation cycles occur leading to the production of ferribiliverdin IXa complex
`[32,33]. The iron of the complex undergoes reduction, which results in the release of free iron and
`biliverdin [31] Then, biliverdin can be further reduced to bilirubin by NADPH-dependent biliverdin
`reductase [34].
`
`2.3. Effect of Exogenous ALA Administration
`
`PpIX is a strong photosensitizer, which assembles in mitochondria of tumor cells leading to their
`damage after light exposure. Although all enzymes involved in the heme biosynthetic pathway are
`necessary, only two of them: ALAS1 and ferrochelatase are considered to be rate-limiting. Normally,
`the activity of ALAS1, is regulated by heme through the negative feedback mechanism. Heme binds to
`the heme-regulatory motif (HRM) in mitochondrial targeting sequence of ALAS and therefore
`prevents transport of ALAS1 precursor to mitochondria [35]. Moreover, there is evidence that heme
`not only regulates ALAS1 mitochondrial import but also attenuates its transcription [36,37]. Normally,
`the feedback mechanism leads to the production of PpIX in such amounts that can be efficiently
`converted to heme by ferrochelatase. Exogenous administration of ALA bypasses the natural
`regulation that heme exerts on ALA synthesis, which leads to increased production of PpIX [7]. The
`efficacy of ferrochelatase is then too low to convert excessively produced PpIX to heme, which results
`in the accumulation of PpIX within cells. About 4-6 hours after administration of ALA, when PpIX is
`already synthesized, target cells are exposed to light, which leads to excitation of the photosensitizer
`and formation of 1O2 that exerts cytotoxic effects.
`
`2.4. Selectivity of PpIX Accumulation in Tumor Tissue in Response to ALA Administration
`
`PpIX has been found to preferentially accumulate in tumor as compared with normal cells. This
`phenomenon can be explained by differences in heme biosynthetic pathway between non-malignant
`and malignant cells. It has been shown that decreased activity of ferrochelatase [38-41] and limited
`availability of iron [42] in tumor cells contribute to increased PpIX accumulation. Enhanced activity of
`enzymes leading to production of PpIX, such as ALAD [43], UROD [43], or PBGD [38,43-44] has
`also been observed in tumor cells.
`
`2.5. Modifications of Heme Biosynthetic Pathway and Its Influence on ALA-PDT
`
`Many studies have been performed in order to better understand the significance of individual parts
`of heme biosynthesis for ALA-PDT. The role of ALAD has been demonstrated by Feuerstein et al.
`who showed that silencing this enzyme with shRNA decreased synthesis of protoporphyrin IX in K562
`
`

`

`Molecules 2011, 16
`
`
`
`4146
`
`erythroleukemic cells and resulted in impaired PDT outcome [45]. Similar effects have been presented
`for inhibition of PBGD with Pb2+ [46]. Furthermore, it has been revealed that pretreatment of LNCaP
`tumor cells with methotrexate significantly improved ALA-PDT efficacy. This effect was related to
`increased synthesis of coproporphyrinogen oxidase, which stimulated the accumulation of PpIX [47].
`Moreover, studies in DBA/2 mice pretreated with PPO inhibitor revealed increased accumulation of
`cytosolic PpIX which potentiated the effect of ALA-PDT [48].
`A significant amount of research has focused on the influence of ferrochelatase activity on the
`efficacy of ALA-PDT. Improved PDT outcome has been shown both after targeted knockdown of
`ferrochelatase by siRNA [49] and after pretreatment of tumor cells with ferrochelatase inhibitors [50].
`Enhancement of PDT effects has been also observed after removal of iron with different iron-chelating
`substances such as desferrioxamine [51] or CP94 [52].
`All these data suggest that efficacy of ALA-PDT is mainly regulated by amount of protoporphyrin
`IX. Therefore, attempts directed either towards its increased production or slower conversion to heme
`seem to be rational approaches to improve the outcome of the therapy.
`
`3. ALA Pharmacokinetics
`
`Although a detailed summary of ALA pharmacokinetics is beyond the scope of this review, several
`issues are briefly outlined here. ALA is usually administrated intravenously (i.v.) or topically. For the
`treatment of bladder cancer ALA is administered intravesically. Moreover, in contrast to other PS, oral
`ALA administration is also feasible. Although this route is preferred by patients, the bioavailability of
`oral ALA is generally lower then after intravenous administration owing to presystemic drug
`elimination [53]. Large biosynthetic PpIX capacity of gastrointestinal mucosal cells and hepatic first
`pass metabolism are two major factors responsible for the reduction of ALA bioavailability. Indeed,
`Dalton et al. observed that only 60% of orally administered ALA is absorbed [54]. Regardless of poor
`oral ALA availability, the tissue PpIX concentrations were comparable after i.v. or oral administration
`[54]. On the other hand, preferential mucosal accumulation of PpIX might be favorable in the
`treatment of tumors of the gastrointestinal tract. Also, limited PpIX accumulation in the underlying
`stroma may reduce damage of deeper layers and risk of perforation or stenosis [55]. In terms of topical
`ALA administration it should be emphasized that due to the fact that stratum corneum is the most
`important barrier for ALA skin penetration, the PpIX fluorescence is observed only at a depth of 0.3 to
`0.6 mm [56-58]. Therefore, a number of studies have focused on ALA derivatization to enhance its
`permeability through the lipid networks in the stratum corneum [59]. Moreover, since hyperkeratosis
`is an important negative factor in ALA uptake pretreatment with keratolytics, curettage/debulking, tape
`stripping, microdermabrasion or laser ablation can be used to improve ALA penetration [60].
`In patients with bladder cancer, the intravesical ALA application results in pharmacokinetic
`advantages in comparison with oral administration. The bladder concentration of ALA is
`approximately 20,000-fold higher in comparison to the systemic circulation. Moreover, only 1% of the
`intravesical dose is absorbed from the bladder by the systemic circulation. Therefore, after intravesical
`ALA administration no systemic phototoxicity should be observed [54].
`Identification of additional factors that affect ALA pharmacokinetics is necessary to design most
`effective ALA-PDT. Changes of numerous parameters of tumor cells as well as their surrounding
`
`

`

`Molecules 2011, 16
`
`
`
`4147
`
`microenvironment may alter PpIX production in malignant lesion. For example, the influence of tissue
`temperature on PpIX accumulation was observed in several studies [61,62]. Other parameters that
`affect PpIX accumulation include pH [63-65], oxygen availability [63,66], illumination conditions, or
`thickness of striatum corneum [67].
`
`4. Modifications of ALA
`
`
`
`Apart from a great potential and numerous advantages of ALA as a PS precursor, a number of
`limitations have also been revealed. ALA has a hydrophilic character and does not penetrate efficiently
`through the skin nor cell membranes. In consequence the production of PpIX after topical ALA
`administration is restricted to a 2–3 mm surface of skin, which might be insufficient to elicit
`satisfactory photosensitization [6]. Moreover, hyperkeratotic lesions drastically diminish ALA
`penetration through the skin. Since the  hydrophilic nature of ALA limits its penetration ability
`restricting the use of ALA-PDT extensive studies were performed in order to facilitate its topical
`delivery. At least 77 various ALA modification and different carriers have been reported [68].
`The most successful ALA derivatives are its esters: methyl ester (methyl aminolevulinate, MAL)
`and hexyl ester (hexyl aminolevulinate, HAL). Elongation of a carbon chain attached to ALA results in
`increased lipophilicity and in consequence higher membrane and skin permeability. The advantage of
`ALA derivatives over ALA can be mainly ascribed to: (i) the rate at which these compounds reach the
`target site, (ii) the rate at which they reach the intracellular space and (iii) the rate of their enzymatic
`conversion into photoactive compounds. Basically, these modifications are favorable and confer
`improved skin penetration. Nevertheless, extensively lipophilic agents tend to accumulate in stratum
`corneum which results in a drop in their biological activity. Different PpIX production profiles as a
`function of drug concentration have been observed for ALA and its esters. ALA and MAL profiles are
`identical, but HAL profile is completely different reaching plateau value at lower concentration [69].
`Research by Lee et al. clearly shows that ALA butenyl, pentenyl and hexenyl esters induce higher
`production of PpIX than ALA or MAL [70]. In order to obtain comparable amount of PpIX with
`ALA-induced porphyrin accumulation, approximately 100 times less concentration of ALA heptyl
`ester is required [71]. On the other hand, undecanoyl-ALA ester, which reveals favorable diffusing
`properties, results in lower PpIX production than ALA itself. Even with the liposomal formulation it
`reaches only standard ALA-mediated PpIX level [72]. The advantage of ALA esters in comparison to
`ALA-inducedPpIX formation is not a general rule, some researchers report that it depends on esterase
`activity which varies within the tissues or cell lines used in the studies [73,74].
`Administration of liposomes entrapping the ALA prodrug into tumor-bearing mice results in
`increment porphyrin biosynthesis as well as higher tumor to normal cells porphyrin ratio [75]. In vitro
`experiments demonstrated lipid sponge forms (thermodynamically stable, amphiphilic liquid) as a
`potential carrier for transdermal drug delivery [76]. It was also suggested that addition of dimethyl
`sulfoxide (DMSO) and ethylenediaminetetraacetic acid (EDTA) to ALA may enhance its delivery to
`hairless mice skin for topical PDT [77]. Conjugating 5-aminolevulinic acid to nanoparticles, including
`biocompatible gold (Au) and chitosan, was one of the solutions suggested to improve ALA delivery to
`the tumor [78,79]. Interestingly, Moan's group proposed topical bioadhesive patch systems that
`enhance selectivity of PpIX accumulation in tumor-bearing mice model [80].
`
`
`

`

`Molecules 2011, 16
`
`
`
`4148
`
`5. Comparison of ALA with Porphyrin-based Photosensitizers
`
`An ideal photosensitizer should be a well-defined chemical compound characterized by a strong
`selective phototoxic effect and an ability to generate active forms of oxygen. Moreover, it should show
`high absorption coefficients in the red part of the electromagnetic spectrum (600–900 nm) allowing the
`light to penetrate the tissue deeply. Its rapid clearance and favorable ADME (absorption, distribution,
`metabolism, excretion) parameters are also of utmost importance. The demand for such a compound is
`a challenge that leads to a synthesis of numerous photosensitizers. They differ from one another in
`terms of chemical structure, which often determines their pharmacokinetics, intracellular localization
`[81] and cytotoxic effect [82]. Generally, the currently used and investigated photosensitizing agents
`can be classified in two groups: porphyrin derivatives and non-porphyrin-based photosensitizers. The
`porphyrin family can be divided into three generations: the first one refers to photosensitizers
`developed between 1970 and 1980 and includes hematoporphyrin derivative (HpD) and Photofrin®.
`The second generation includes porphyrin derivatives that were intended to overcome the limitations
`of the former and the third generation encompasses photosensitizing agents conjugated with antibodies
`and other biological targeting molecules. So far only five photosensitizers have been approved for
`clinical PDT – porfimer sodium (Photofrin®), 5-aminolevulinic acid (ALA, Levulan®), and its methyl
`ester MAL (Metvix®), temoporfin (Foscan®), verteporfin (Visudyne®) and talaporfin (Laserphyrin®;
`the latter only in Japan). ALA is the only photosensitizer registered for topical use, particularly useful
`for the local treatment of superficial skin lesions. ALA-PDT has demonstrated high efficacy, minimal
`side effects and excellent cosmetic effects in a wide range of benign and malignant conditions.
`ALA has several advantages over other photosensitizers, namely rapid metabolism and high
`selectivity for malignant lesions. Prompt systemic clearance of ALA-induced PpIX within 24 h
`eliminates prolonged photosensitivity and allows treatment to be repeated at regular intervals (as
`frequently as every 48 h) without cumulative effects and risk of damage to normal tissue. Nevertheless,
`significant disadvantages of ALA-PDT include pain associated with treatment [83], limited depth of
`tumor penetration [84,85], as well as individual variations among patients that affect ALA absorption
`and pharmacokinetics that influence effective ALA concentrations in the treated area [86]. Moreover,
`ALA-PDT appeared to be less efficient in destroying cutaneous lesions when compared with
`Photofrin-PDT [86]. A general comparison of ALA and different porphyrin-based photosensitizers,
`taking into account their physical and chemical properties as well as molecular mechanisms associated
`with the influence on tumor cells is provided in Table 1.
`
`Chemical
`classification
`Hematoporphyrin
`
`Table 1. Properties of selected photosensitizers.
`Photosensitizer/trade
`Clinical approval/
`Cellular
`name/company
`clinical trials
`localization
`Approved
`Plasma
`Porfimer sodium -
`membrane
`combination of monomers,
`Golgi
`dimers and oligomers of
`apparatus
`hematoporphyrin
`[139,140]
`derivative (around 85%
`oligomeric material and
`mixture of more than 60
`compounds)/ Photofrin
`/Axcan Pharma, QLT
`Pharmaceuticals
`
`Advantages
`Most commonly
`used photo-
`sensitizer, the
`longest clinical
`history and
`patient record,
`pain-free
`treatment
`
`Disadvantages
`Complex composition,
`slow clearance rate,
`prolonged
`photosensitivity up to 3
`months, low
`fluorescence quantum
`yield, low efficiency in
`ROS generation,
`limited penetration and
`efficacy in deep and
`bulky tumors
`
`

`

`Molecules 2011, 16
`
`
`
`Protoporhyrin
`
`Pro drug (5-aminolevulinic
`acid - ALA) converted to
`photoactive protoporhyrin
`IX/ Levulan, Levulan
`Kerastick (for topical use)/
`Dusa Pharmaceuticals)
`
`Table 1. Cont. 
`Approved
`Mitochondria,
`cell
`membranes,
`cytosol,
`cytosolic
`membranes
`[141]
`
`
`
`Texaphyrins
`
`Porphycenes
`
`Aminolevulinic esters/
`Metvixia/Galderma
`Benzvix, Hervix used for
`photodiagnosis [142]
`(PhotoCure AS)
`Motexafin lutetium/
`Lutrin, Optrin, Antrin/
`Pharmacyclics
`
`
`Various porphycene
`derivatives, modifiable
`isomers of porphyrin/ NDA
`
`Approved
`
`
`
`Completed clinical
`trials phase I
`
`Phase II clinical trial
`of topical ATMPn
`(9-acetoxy-
`2,7,12,17-tetrakis(β-
`methoxyethyl)-
`porphycene)
`
`Primarily in
`lysosomal
`compartment
`[143]
`
`Mitochondria
`Lysosomes
`ER
`plasma
`membrane
`[144]
`
`Purpurins
`
`Tin etyl etiopurpuryn
`rostaporfin/ SnET2,
`Photrex, Purlytin/ Miravant
`Medical Technologies
`
`In clinical trials
`phase III
`
`
`Mitochondria
`lysosomes
`
`Pheophorbides
`
`WST-09 (padoporfin,
`palladium
`bacteriopherophorbide
`/Tookad and WST-
`11(padeliporphin) /Stakel/
`Steba Biotech
`
`WST11- phase I and
`II, WST-09 – phase
`II
`
`NDA
`
`4149
`
`Pain associated with
`treatment (need of
`local analgesia), need
`of prolonged contact
`period before
`illumination
`
`Painful treatment
`
`
`Severe pain during
`the phototherapy
`(need of local
`anesthesia)
`
`Photobiological
`properties still poorly
`explored
`
`Post-treatment pain,
`long-lasting photo-
`sensitization up to 14
`days [146], poor
`stability in water
`(need of formulation
`in lipid emulsions,
`which can lead to
`allergic reactions)
`Narrow time window
`available for light
`delivery (important
`in clinical settings)
`
`Easy synthesis
`and formulation,
`minimal
`photosensitivity
`for no more than
`24 hours (rapid
`clearance),
`excellent
`cosmetic results,
`especially eyelids,
`inexpensive,
`possibility of
`application
`without doctor’s
`supervision (oral
`and topical
`administration),
`can be
`administered at
`regular intervals
`(even every 48h),
`high selectivity
`due to
`metabolism of
`ALA in malignant
`cells and
`pilosebaceous
`units
`Improved skin
`penetration -
`greater selectivity
`
`Deep tissue
`penetration
`
`Efficient ROS
`generation,
`possibility of
`various structural
`and chemical
`modifications that
`improve half-life
`and
`enhance
`therapeutic
`efficiency
`Excellent
`cosmetics effect,
`effective in
`treatment of
`locally advanced
`metastatic
`malignancies
`[145]
`
`Little or no skin-
`associated
`sensitivity,
`greater tissue
`penetration [84],
`the possibility of
`repeated
`treatments
`
`

`

`Molecules 2011, 16
`
`
`
`Chlorins
`
`Talaporfin sodium, mono-
`L-aspartyl chlorine 6/
`NPe6, MACE, LS11,
`Laserphyrin, Photolon,
`Aptocine/
`Light Sciences Oncology
`
`Table 1. Cont. 
`Approved
`Lysosomes
`
`
`
`
`
`
`Phthalocyanines
`
`
`Approved
`
`
`ER
`mitochondria
`
`Temoporfin, meta-
`tetrahydroxyphenylchlorine,
`mTHPC/
`Foscan/
` Biolitec Pharma
`
`
`HPPH 2-(1-hexyloxyethyl)-
`2-devinyl pyropheo-
`phorbide/ Photochlor/
`Roswell Park Cancer
`Institute
`Aluminium (III) phthalo-
`cyanine tetrasulphate,
`AlPcS4/Photosens (mixture
`of sulfonated aluminium
`phtalocyanines)/
`developed in Russia,
`General Physics Institute
`
`Mitochondria
`
`Mitochondria
`
`In naturally occurring
`veterinary tumors
`(cats and dogs),
`clinical trials phase I
`and II
`In naturally occurring
`veterinary tumors,
`several clinical trials
`in Russia
`
`
`
`Silicon phthalocyanine 4/
`Pc4/ Case Western Reserve
`University
`
`Ongoing clinical
`trials phase I
`
`Mitochondria
`ER
`
`4150
`
`In clinically needed
`high doses little
`selectivity to tumor
`tissue occurred
`[147]
`
`Accumulation in the
`skin, requires strict
`protection of the
`eyes and skin from
`sunlight for up to 6
`weeks, long drug-
`illumination
`interval, requires
`very precise
`illumination
`(reflected light can
`produce
`photodynamic
`reaction) and
`accurate dosimetry
`Phototoxicity not
`determined in higher
`doses
`
`Problems with
`purification,
`typically final
`product is a mixture
`of mono- di- tri- and
`tetrasulphonated
`derivatives, in water
`aggregate at
`relatively low
`concentrations
`which results in loss
`of photo-chemical
`activity
`Heterogeneous
`distribution within
`and between lesions
`detected by
`noninvasive
`spectroscopy [148]
`
`Excellent singlet
`oxygen yield, used
`in Litx therapy
`(Light Infusion
`Technology),
`where talaporfin is
`illuminated for
`prolonged time (1-
`3 h) locally with
`light-emitting
`diodes (LEDs)
`implanted in the
`tumor, minimal
`skin photo-
`sensitivity
`High singlet
`oxygen yield (low
`drug dose and low
`light dose -
`20J/cm2 are
`required to
`generate
`photodynamic
`reaction), low
`activation energy
`and short time
`treatment, long
`half-life in triplet
`state
`
`Minimal sunlight
`photosensitivity,
`relatively easy to
`synthesize
`
`High singlet
`oxygen yield with
`long-lived triplet
`states due to
`presence of
`aluminum atom,
`due to enhance
`fluorescence can
`be used for
`diagnostic
`purpose, minimal
`photosensitivity
`
`High singlet
`oxygen yield with
`long-lived triplet
`states due to
`presence of silicon
`atom, good
`efficacy in both
`preclinical and
`clinical studies,
`due to enhance
`fluorescence can
`be used for
`diagnostic
`purposes
`
`

`

`Molecules 2011, 16
`
`
`
`4151
`
`Benzoporphyrins Verteprofin/Visudyne
`/Novartis
`
`Table 1. Cont.
`Approved
`Mitochondria
`and ER
`
`Deep tissue
`penetration,
`minimal photo-
`sensitization up to
`48 h, effectiveness
`in neovascular
`lesions, successful
`in cutaneous
`lesions
`Abbreviations used: ER – endoplasmatic reticulum, NDA – no data available, ROS – reactiv

This document is available on Docket Alarm but you must sign up to view it.


Or .

Accessing this document will incur an additional charge of $.

After purchase, you can access this document again without charge.

Accept $ Charge
throbber

Still Working On It

This document is taking longer than usual to download. This can happen if we need to contact the court directly to obtain the document and their servers are running slowly.

Give it another minute or two to complete, and then try the refresh button.

throbber

A few More Minutes ... Still Working

It can take up to 5 minutes for us to download a document if the court servers are running slowly.

Thank you for your continued patience.

This document could not be displayed.

We could not find this document within its docket. Please go back to the docket page and check the link. If that does not work, go back to the docket and refresh it to pull the newest information.

Your account does not support viewing this document.

You need a Paid Account to view this document. Click here to change your account type.

Your account does not support viewing this document.

Set your membership status to view this document.

With a Docket Alarm membership, you'll get a whole lot more, including:

  • Up-to-date information for this case.
  • Email alerts whenever there is an update.
  • Full text search for other cases.
  • Get email alerts whenever a new case matches your search.

Become a Member

One Moment Please

The filing “” is large (MB) and is being downloaded.

Please refresh this page in a few minutes to see if the filing has been downloaded. The filing will also be emailed to you when the download completes.

Your document is on its way!

If you do not receive the document in five minutes, contact support at support@docketalarm.com.

Sealed Document

We are unable to display this document, it may be under a court ordered seal.

If you have proper credentials to access the file, you may proceed directly to the court's system using your government issued username and password.


Access Government Site

We are redirecting you
to a mobile optimized page.





Document Unreadable or Corrupt

Refresh this Document
Go to the Docket

We are unable to display this document.

Refresh this Document
Go to the Docket