Open access

The Menace of Melanoma: A Photodynamic Approach to Adjunctive Cancer Therapy

Written By

L.M. Davids and B. Kleemann

Submitted: 31 August 2012 Published: 30 January 2013

DOI: 10.5772/53676

From the Edited Volume

Melanoma - From Early Detection to Treatment

Edited by Guy Huynh Thien Duc

Chapter metrics overview

2,943 Chapter Downloads

View Full Metrics

1. Introduction

Metastatic malignant melanoma (MMM) remains one of the most dreaded skin cancers worldwide. Numerous factors contribute to its resistance to hosts of treatment regimes and despite significant scientific advances over the last decade in the field of chemotherapeutics and melanocytic targets, there still remains the need for improved therapeutic modalities. Photodynamic therapy (PDT), a minimally invasive therapeutic modality has been shown to be effective in a number of oncologic and non-oncologic conditions. Using second-generation stable, lipophillic photosensitizers with optimised activation wavelengths, PDT may be a promising tool for adjuvant therapy and even pre-treatment in combating melanoma. Potential targets for PDT in melanoma eradication include cell proliferation inhibition, activation of cell death and reduction in pro-survival autophagy, a decrease in the cellular melanocytic antioxidant system and a disruption in the endogenous multi-drug resistant (MDR) cellular machinery. This chapter highlights the current knowledge with respect to these characteristics and suggests that PDT be considered as a good candidate for adjuvant treatment in post-resected malignant metastatic melanoma. Furthermore, it suggests that primary consideration must be given to organelle-specific destruction in melanoma specifically targeting the melanosomes – the one organelle that is specific to cells of the melanocytic lineage that houses the toxic compound, melanin. We believe that using this combined knowledge may eventually lead to an effective therapeutic tool to combat this highly intractable disease.

1.1. Melanoma clinical statistics

Melanoma accounts for 4% of all dermatologic cancers but remains responsible for 80% of deaths from skin cancer with the average patient diagnosed with disseminated metastases surviving for an average of 5 years (Cancer facts and figures, 2003, Atlanta, American Cancer Society, 2003]. According to the World Health Organization (WHO) melanoma skin cancer has been increasing over the past decades with a global estimation of 132 000 melanoma-related skin cancers reported to occur each year. Over the past 50 years, melanoma incidence has risen by 3–8% per year in most people of European background, with the greatest increases in elderly men [1]. In Europe, the current estimates at 15-20 per 100 000 people predominating in the 20-35 year old age group in Caucasians [2]. South Africa, next to Australia, has one of the highest incidences of malignant melanoma in the world. Reliable statistics for South Africa are lacking, however currently an estimate figure for the South African Cape region is 69 new cases per year per population of 100 000 Caucasians (Australia is 65 per 100 000). This means that 1 in 1429 people will develop malignant melanoma. The age-standardised incidence of melanoma was 27.2 per 100 000 for males and 22.2 per 100 000 for females from 1990-1999 but this increased to 36.9 for males and 33.5 per 100 000 for females (2000-2003) (CANSA association of South Africa www.melanoma.co.za/D_doccnr_MFS.asp) (Table 1).

Age-standardised incidence (105 /yr) Lifetime risk (incidence) Incidence trend over 10 years Mortality trend over 10 years Most common cancer (ranking)
Australia (2001)
Men
Women
41.4 (world)
31.1 (world)
1 in 25
1 in 35
22% increase
12% increase
2% increase
0% increase
4th
3rd
South Africa (2000)
Men
Women
36.9 (world)
33.5 (world)
1 in 29
1 in 40
33% increase
27% increase
1.5% increase
1% increase
4th
3rd
USA (2001)
Men
Women
21.4 (world)
13.8 (world)
1 in 53
1 in 78
31% increase
25% increase
0% increase
1% decrease
5th
7th
UK (2000)
Men
Women
9.7 (world)
11.2 (world)
1 in 147
1 in 117
59% increase
41% incraese
20% increase
3% increase
12th
7th

Table 1.

Melanoma statistics in 2 southern and northern hemisphere countries

Despite extensive research and clinical trials, the prognosis and survival of metastatic melanoma remains dismal. Early detection of localized melanoma may be cured through surgery however there is no therapy for metastatic melanoma or melanoma with metastatic potential. In addition, recurrence rates of resected melanoma remain high. Because melanoma is inherently resistant to traditional forms of chemotherapy and radiotherapy [3], various strategies have been developed for treatments which include immunotherapy eg. interleukin-2 (IL-2) [4], radiotherapy [5] biochemotherapy [6-9] and gene therapy [4,10]. A limited number of these therapies have progressed to human clinical trials but their outcomes remain negligible. One promising therapy is high-dose interferon (IFN) alpha-2b therapy which has just recently been approved as the only adjuvant therapy for melanoma approved by the US Food and Drug Administration [11]. The other is the use of BRAF kinase inhibitors such as vemurafenib [12]. Despite convincing evidence of improved disease-free survival associated with this therapy, the overall survival remains negligible or very small [13-15]. In addition, a number of melanoma-specific and melanoma-associated tumor antigens such as gp100, MART-1 and MAGE3 have been cloned [16] and the hope is that these potential antigens may be developed to stimulate tumor-specific T cells to eliminate melanoma cells [17]. Despite these advances, there remains the need for the development of novel and effective approaches to treat melanoma and this review explores the possibility of using photodynamic therapy (PDT) as an adjuvant therapy alone or in combination with current therapeutics to combat melanoma.

1.2. Melanoma origins

Melanoma represents the malignant phenotype of a skin melanocyte. Melanoma occurs most frequently after intermittent exposure to UV radiation and in people with chronic sunburns. Epidemiologic data suggest that chronic or low-grade exposures to UV induce protection against DNA damage, whereas acute, intense UV exposure leads to DNA damage and concomitant genetic alterations in the melanocyte genome [18]. It develops as a result of accumulated abnormalities in genetic pathways within the melanocyte which give way to increased cell proliferation and prevent normal pathways of apoptosis in response to DNA damage. Furthermore, this damage results in the selection for genetic mutations that allow all aspects of the malignant phenotype, including stimulation of blood vessel growth, evasion of the immune response, tumour invasion, and metastasis [19]. Although the mechanisms of differential cancer cell killing are poorly understood [20], selection of cells that are resistant to apoptotic mechanisms might contribute to the resistance of melanoma cells to the cytotoxic effects of chemotherapy, radiotherapy, and immunotherapy, especially through the expression of apoptosis inhibitors such as B-cell lymphoma derived protein 2 (Bcl-2) and BclxL [21].

Melanocytes progress through a series of steps toward malignant transformation by the acquisition of various phenotypic features. The particular histological features characterising each step of progression are the visible manifestations of underlying genetic changes [22]. Originating from a benign nevus, melanocytes undergo aberrant growth within the lesion subsequently displaying irregular borders, a change in colour and often an associated allergic response. At this stage the lesion is considered dysplastic. At a molecular level, these changes are associated with abnormal activation of the mitogen-activated protein kinase (MAPK) signalling pathway resulting in somatic mutations in the N-RAS and BRAF genes which are associated with about 15 and 50% of melanomas, respectively [23,24]. There is complementarity between the presence of NRAS and BRAF mutations in any individual melanoma since each has the same effect of causing unrestrained cell proliferation.

In addition, mutations in both the cyclin-dependent kinase inhibitor 2A (CDKN2A) and the phosphatase and tensin homologue (PTEN) gene increases the probability of dysplastic naevi becoming malignant [25]. This genetic locus is frequently targeted for disruption in melanomas [26]. When defective, p16 is unable to inactivate CDK4 and CDK6, which phosphorylate Rb, releasing the transcription factor E2F and leading to cell cycle progression [27].The molecule that is usually central to protection against DNA damage, p53, is rarely mutated early in melanoma, which is possibly one of several adaptations to permit survival of cells responsible for generating sun-protective pigment, melanin [28]. Interestingly, by-products of melanin biosynthesis can themselves cause oxidative stress and contribute to malignant change.

Further progression of melanoma is associated with decreased differentiation and clonal proliferation leading to the radial growth phase (RGP). Clinically, RGP presents as patches or plaques which can measure up to 2.5cm. Superficial spreading melanoma lesions are slightly raised and show striking variations of red, blue, white, brown, and black coloration. In RGP, melanoma mitoses are frequently seen in the epidermis but rarely in the dermis. After complete surgical excision of the tumor, RGP melanomas are usually associated with longterm metastasis-free survival [29-33]. RGP cells can progress to vertical growth phase (VGP) cells which breach the basement membrane and invade the dermis as nodules or nests of cells. Vertical growth phase (VGP) melanomas usually present as gray-black, blue-black, or even amelanotic nodules. In late or developed VGP, melanomas form expansile nodules in the dermis with cytology different from melanoma cells in the overlying epidermis. Mitotic figures are variably present, and tumor aggregates may extend into the reticular dermis or even subcutaneous fat. Dermal tumoral nests are larger in VGP than in RGP. Moreover, these cells are considered to have metastatic potential. Interestingly, not all melanomas pass through each of these individual phases – RGP and VGP can both develop directly from melanocytes or naevi and both can progress directly to metastatic malignant melanoma [34]. Moreover, the transition from RGP to VGP in cutaneous melanoma isassociated with the loss of c-KIT expression and the gain of the melanoma cell adhesion molecule (MCAM/MUC18) [35].

Increased proliferation and survival, chemoresistance, the ability to resist apoptosis, the induction of autophagy and the presence of the pigment melanin have all been listed as reasons contributing to the high mortality rates associated with cutaneous melanoma. Each of these topics will be dealt with in the context of targeting them with PDT.

1.3. Photodynamic Therapy (PDT) as a cancer treatment

PDT is a minimally invasive therapeutic modality which has been shown to be effective in several types of cancer including non-melanoma skin cancer (NMSC) and other skin tumors such as lymphoma as well as non-oncological conditions such as psoriasis vulgaris, acne vulgaris and human papilloma virus-induced skin disease [36,37]. The basis of PDT is the systemic or topical application and preferential uptake of a photosensitizer (PS). The PS is then activated at a specific wavelength of light and in the presence of oxygen, produces reactive oxygen species (ROS). The accumulative presence of these cytotoxic photoproducts start a cascade of molecular and biochemical events resulting in cell death via apoptotic or necrotic mechanisms [38,39].

The main advantage of PDT over conventional cancer treatments are i) it has a very low systemic cumulative toxicity allowing repeated dosing, ii) its ability to destroy tumors selectively (this seems to be related to the lipophillic nature of photosensitizers). Due to this selectivity, damage to normal surrounding cells is minimal. Finally, iii) PDT can be applied alone or in combination as an adjuvant therapeutic modality with chemotherapy, surgery, radiotherapy and immunotherapy [40,41]. These properties have led to PDT receiving increased support from preclinical research [42,43]. PDT requires three elements to be efficacious - a good PS, a coherent light source and the presence of molecular oxygen. A large amount of data with regard to these three elements over the last few years have resulted in the development of more naturally-derived, efficacious, second-generation photosensitizers.

1.3.1. Photosensitizers and melanoma-PDT

Photosensitizers are critical to the successful eradication of malignant cells and numerous first and second-generation photosensitizers have been tested both clinically (in vivo) and in vitro over the past years (for a detailed summary of melanoma-PDT research see Table 2). The structure of many PS is based on the tetrapyrrol ring eg. protoporphyrin IX, Photofrin and chlorines related to it eg. phthalocyanines. Newer, more stable second-generation PS include natural hydroxyquinone chromophores such as hypericins and porphycenes [44-47]. It is now accepted that a good PS for PDT is – i) chemically pure with good stability, ii) preferentially accumulated and retained by target tissue, iii) minimal toxicity in the absence of light with maximal efficacy upon activation, iv) high quantum yield of 1O2 with an associated high molecular extinction coefficient [40]. Due to these properties, a number of synthetic or natural compounds have thus far been studied for a variety of cancers however these have been limited to porphycenes (structural isomers of porphyrins) such as aminolevulinic acid (ALA, trade name, Levulan®) and methylaminolevulinic acid (MAL, trade name, Metvix® ) for the treatment of squamous cell (SCC) and basal cell carcinomas (BCC) as well as actinic keratoses [48-50] (Table 2).

Type of study Tumour/Cell line Photosensitizer Ref
in vitro B16-F10 melanoma cells magnetoliposomes (MLs) loaded with zinc phthalocyanine (ZnPc) complexed with cucurbituril (CB) (CB:ZnPc-MLs) [51]
in vivo mice carrying B16-F10 melanoma xenografts butadiyne-linked conjugated porphyrin dimer (Oxdime) [52]
in vivo subcutaneous amelanotic melanoma transplanted in C57/BL6 mice pheophorbide a (Pba) and monomethoxy-polyethylene glycol-Pba [53]
in vitro A375, UCT Mel-1 human melanoma cells hypericin and kojic acid (depigmenting agent) [54]
in vitro/ in vivo B16F10 mouse melanoma cells and lung melanomas in C57BL/6 mice aminolevulic acid, gaussia luciferase, and its' substrate coelenterazine; murine neural stem cells (NSCs) and rat umbilical cord matrix-derived stem cells (RUCMSCs) with a plasmid expressing gaussia luciferase [55]
in vitro C32 human melanoma cells Ficus carica L. cultivar Dottato extracts [56]
in vitro Melanoma, keratinocyte and fibroblast cells aluminum tetrasulfophthalocyanines [57]
in vivo B16-F1 and Cloudman S9 melanoma-bearing mice chlorin e(6) and modular nanotransporters targeted to α-melanocyte-stimulating hormone (αMSH) and epidermal growth factor (EGF) receptor [58]
in vivo malignant melanoma mouse model methylene blue [59]
in vitro A549 and S91 melanoma cells halogenated sulfonamide bacteriochlorins [60]
in vitro A375 melanoma cells carotenoids (neoxanthin, fucoxanthin and siphonaxanthin) [61]
in vitro melanoma cells 2 cationic octanuclear metalla-cubes dual photosensitizers and chemotherapeutics [62]
in vitro A375 melanoma cells Cachrys pungens Jan extracts from Italy [63]
in vitro B16F10 murine melanoma indocyanine green (ICG) and hyperthermia [64]
in vitro B78-H1 murine melanoma cells pheophorbide a [65]
in vitro S91 Cloudman melanoma cells and DBA mice synthetic chlorin derivative (TCPCSO₃H) [66]
in vitro/ in vivo Melanoma cells and xenograft melanoma model cis-Dichlorobis [3,4,7,8-tetramethyl-1,10-phenanthroline) rhodium(III) chloride (OCTBP) [67]
in vitro M21 human melanoma cells Hedyotis corymbosa extracts [68]
in vitro A375, UCT Mel-1 human melanoma cells hypericin and phenylthiourea (depigmenting agent) [69]
in vitro melanoma, keratinocyte and fibroblast cells zinc tetrasulfophthalocyanines (ZnTSPc) [70]
in vitro melanoma cells PDT and Lycopene, β-carotene, vitamin C, N-acetylcysteine, trolox, N-tert-butyl-α-phenylnitrone and HO-1 activity inhibitor zinc protoporphyrine IX (ZnPPIX) [71]
in vivo mice bearing mouse melanomas verteporfin [72]
in vitro/ in vivo S91 mouse melanoma cells and DBA mice 5,10,15,20-tetrakis[2-chloro-5-sulfophenyl)bacteriochlorin (TCPBSO3H) [66]
in vitro WM451LU melanoma cells photosensitizers and heme oxygenase I (HO-I) and poly(ADP-ribose) polymerase (PARP) inhibitors [73]
in vitro A375 melanoma cells 5-aminolevulinic acid [5-ALA) and novel metallophthalocyanine (MPc) [74]
in vivo transplanted B16 melanoma novel derivatives of chlorin e[6] [75]
in vitro/ in vivo melanoma bearing mice C(60)-(Glc)1 (D-glucose residue pendant fullerene) and C(60)-(6Glc)1 (a maltohexaose residue pendant fullerene) [76]
in vitro SK-MEL-188 (human melanoma) cells chlorin and bacteriochlorin derivatives of 5,10,15,20-tetrakis[2-chloro-5-sulfophenyl)porphyrin [77]
in vitro B16 melanoma cells IPL and IPL plus 5-ALA [78]
in vitro melanoma cells bacteriochlorins and photofrin [79]
in vitro C57 mice bearing a sub-cutaneously transplanted melanoma Zn(II)-phthalocyanine disulphide (C11Pc) [80]
in vitro WM 1552C human melanoma cells liposomes (LP) and nanocapsules (NC) containing Chloroaluminum phthalocyanine (CIAIPc) [81]
in vitro A375 melanoma cells 5,15-Diarylporphyrins (1-5) and Photofrin [82]
in vivo B16 melanoma tumours on mice 2 doses of photosensitizer [83]
in vitro B16 mouse melanoma cells chlordiazepoxide (CDZ) [84]
in vitro/ in vivo B78H1 amelanotic mouse melanoma cells and C57BL/6 mice bearing a subcutaneously transplanted B78H1 amelanotic melanoma. octabutoxy-naphthalocyanines [85]
in vitro G361 human melanoma cells zinc-5,10,15,20-tetrakis(4-sulphonatophenyl) porphyrine (ZnTPPS(4)), chloraluminium phtalocyanine disulfonate (ClAlPcS(2)) and 5-aminolevulinic acid (ALA) [86]
in vitro A375 human melanoma cells B16F10 mouse melanoma cells Pc4 encapsulated in silica nanoparticles [87]
in vitro Sk-Mel-28 human skin melanoma cells indocyanine green [88]
in vitro UCT Mel-1 and A375 human melanoma cells hypericin [89]
in vitro G361 human melanoma cells chloroaluminum phthalocyanine (ClAlPc) and ultrasound [90]
in vitro A375 human melanoma cells 5-aminolevulinic acid (ALA) [91]
in vitro/ in vivo B16F1 mouse melanoma cells and C57BL6 mice bearing a subcutaneously injected B16F1 melanoma. methylene blue [92]
in vitro/ in vivo B16F1 mouse melanoma cells and C57BL6 mice bearing a subcutaneously injected B16F1 melanoma. carboranyl-containing chlorin
(TPFC)
[93]
in vitro/ in vivo human malignant melanoma cells (MMCs) porfimer sodium [94]
in vivo B57BL/6 mice bearing a B16BL6 melanoma porfimer sodium and antibodies neutralizing decay-accelerating factor (DAF), complement-receptor-1-related protein y (Crry), and protectin [95]
in vitro G361 human melanoma cells porphyrines (TPPS4, ZnTPPS4 and PdTPPS4 [96]
in vitro/ in vivo B-16 mouse melanoma cells and subcutaneous B-16 melanoma-bearing C57BL/6 mice 5,10,15,20-tetraphenylporphin-loaded PEG-PE micelles [97]
in vitro UCT Mel-1 and UCT Mel-3 human melanoma cells hypericin [98]
in vitro Me300 human melanoma cells Five 5,10,15,20-tetra[4-pyridyl)porphyrin (TPP) areneruthenium(II) derivatives and a p-cymeneosmium and two pentamethylcyclopentadienyliridium and -rhodium analogues [99]
in vitro B16F10 melanotic melanomas transplanted to nude mice methyl 5-aminolevulinate (MAL) and depigmentation with violet light [100]
in vitro S-91 mouse melanoma cells titanium dioxide modified with platinum(IV) chloride complexes (TiO2/PtCl4) [101]
in vitro G361 human melanoma cells zinc-5,10,15,20-tetrakis(4-sulphonatophenyl) porphyrine (ZnTPPS4) and atomic force microscopy [102]
in vitro WM451Lu metastatic human melanoma cells 5-aminolevulinic acid (ALA) [103]
in vitro B16F1 mouse melanoma cells meso-tetra[4-nido-carboranylphenyl)porphyrin (H2TCP) [104]
in vitro B16F10 mouse melanoma cells solketal-substituted phthalocyanine (Si(sol)2Pc in mPEG-b-p(HPMAm-Lac2) micelles
[105]
in vitro B19 mouse and G361 human melanoma cells phthalocyanine ClAlPcS(2) [106]
in vitro S91 mouse and SKMEL 188 human melanoma cells 5,10,15,20-tetrakis[2-chloro-3-sulfophenyl)porphyrin (TCPPSO(3)H), [107]
in vivo choroidal melanomas in 46 New Zealand albino rabbit eyes hematoporphyrin monomethyl ether [108]
in vitro G361 human melanoma cells 3 porphyrin sensitizers (TPPS(4), ZnTPPS[4] and PdTPPS(4)) [109]
in vitro YUSAC2/T34A-C4 human melanoma cell line porfimer sodium [110]
in vivo subcutaneous B16BL6 melanoma-bearing C57BL/6 mice BPD, ce6, Photofrin, and mTHPC and gamma-inulin [111]
in vitro A375 human melanoma cells acridine orange [112]
in vitro Cloudman S91/I3 mouse melanoma cells photofrin II (PfII- porfirmer sodium), verteporfin, and merocyanine 540 (MC540) [113]
in vivo A-Mel-3 melanomas implanted in the dorsal skin fold chamber of Syrian Golden hamsters 5-aminolaevulinic acid (ALA) [114]
in vivo B-16 melanoma-bearing C57BL/6 mice ATX-S10 No (II) and intratumoral injection of naïve dendritic cells (IT-DC) [115]
in vitro/ in vivo B16F1 mouse melanoma cells and C57BL6 mice bearing a subcutaneously injected B16F1 melanoma. Zn(ii)-phthalocyanine derivative bearing four 10B-enriched o-carboranyl units [10B-ZnB4Pc) [116]
in vitro human Beidegröm Melanoma (BM) cell line porfirmer sodium (photofrin II) [117]
in vitro G361 human melanoma cells ZnTPPS(4) sensitizer bound to cyclodextrin hpbetaCD [118].
in vitro B78H1 mouse melanoma cells Ni(II)-octabutoxy-naphthalocyanine (NiNc) [119]
in vitro G361 human melanoma cells ZnTPPS(4) sensitizer bound to cyclodextrin hpbetaCD [120]
in vitro M2R mouse melanoma cells O-[Pd-bacteriochlorophyllide]-serine methyl ester (Pd-Bchl-Ser) [121]
in vitro B78H1 melanoma cells liposome-delivered Ni(II)-octabutoxy-naphthalocyanine [119]
in vitro human choroidal melanoma (CM) cells tetrahydroporphyrin tetratosylat (THPTS) [122]
in vivo M2R mouse melanoma xenografts WST11 [123]
in vitro B16 mouse melanoma cells 5-aminolevulinic acid [5-ALA) ester derivatives [124]
in vivo B-16 melanoma-bearing C57BL/6 mice metal-free sulfonated phthalocyanine (H(2)PcS(2.4)) [125]
in vitro A375 human melanoma cells alpha-methylene-gamma-butyrolactone-psoralen heterodimer 2 [126]
in vitro B16 mouse melanoma cells 5-aminolevulinic acid (ALA) [127]
in vivo C57BL6 mice bearing a subcutaneously injected B16F10 melanoma silkworm excreta (SPbalpha) porfirmer sodium [128]
in vitro M3Dau human melanoma cells silicon-phthalocyanines (SiPc) and chloro-aluminium Pc (ClAlPc), [129]
in vitro Me45 human melanoma cells meso-tetra-4-N-methylpyridyl-porphyrin iodide and 5,10-di-[4-acetamidophenyl)-15,20-di-[4-N-methylpyridyl) porphyrin [130]
in vitro G361 human melanoma cells meso-tetrakis[4-sulphonatophenyl)porphine (TPPS4) and zinc metallocomplex (ZnTPPS4) [131]
in vitro G361 human melanoma cells ATX-S10(Na) [132]
in vivo syrian Golden hamsters fitted with dorsal skinfold chambers containing A-Mel-3 melanoma cells 5-aminolaevulinic acid (ALA) [133]
in vitro B16 mouse melanoma cells 5-aminolaevulinic acid (ALA) [134]
in vivo UB900518 human melanoma cells transplanted on nude (nu/nu) CD-1 mice Liposomal meso-tetrakis-phenylporphyrin (TPP) [135]
in vivo pigmented choroidal melanoma 44 New Zealand albino rabbit eyes liposomal preparation of benzoporphyrin derivative (BPD), verteporfin [136]
in vitro S91 mouse and SKMEL 188 human melanoma cells indocyanine green (ICG) [137]
in vitro B16A45 (B16) mouse melanoma cells delta-aminolevulinic acid (ALA) and meta(tetrahydroxyphenyl)chlorin or m-THPC [138]
in vitro B16 mouse melanoma cells m-THPC and four apoptosis inhibitors: BAPTA-AM, Forskolin, DSF, and Z.VAD.fmk [47]
in vitro Bro, SKMel-23, SKMel-28 5-aminolevulinic acid (ALA) [139]
In vitro SKMEL 188 human melanoma cells tritolylporphyrin dimer (T-D). [140]
in vivo Syrian Golden hamsters fitted with dorsal skinfold chambers containing A-Mel-3 melanoma cells 5-aminolaevulinic acid (ALA) [141]
in vivo Nude CD1 mice bearing malignant M2R melanoma xenografts bacteriochlorophyll-serine (Bchl-Ser), [142]
in vitro SK-23 mouse melanoma and SK-Mel 28 human melanoma methylene blue [143]
in vitro B78H1 melanoma cells liposome-incorporated Ni(II)-octabutoxy-naphthalocyanine (NiNc), [144]
in vitro B78H1 melanoma cells Cu(II)-hematoporphyrin (CuHp) [145]
in vivo Syrian Golden hamsters fitted with dorsal skinfold chambers containing A-Mel-3 melanoma cells 5-aminolaevulinic acid (ALA) [146]
in vitro M6 human melanoma cells dichlorosilicon phthalocyanine (Cl2SiPc) and bis(tri-n-hexylsiloxy) silicon phthalocyanine (HexSiPc) [147]
in vivo C57/BL6 mice bearing a subcutaneously transplanted B1 melanoma benzoporphyrin derivative monoacid ring A (verteporfin, BPD-MA) [148]
in vitro SkMel-23 melanoma cells 5-aminolaevulinic acid (ALA) [149]
in vivo C57/BL6 mice bearing a subcutaneously transplanted B16 melanoma Si(i.v.)-naphthalocyanine (isoBO-SiNc) [150]
in vivo C57/BL6 mice bearing a subcutaneously transplanted B16 melanoma aluminum phthalocyanine (AlpcS4 [151]
in vivo C57/BL6 mice bearing a subcutaneously transplanted B16F10 melanoma lutetium texaphyrin (PCI-0123), [152]
in vivo Syrian Golden hamsters fitted with dorsal skinfold chambers containing A-Mel-3 melanoma cells 9-acetoxy-2,7,12,17-tetrakis-(beta-methoxyethyl)-porphycene (ATMPn) [153].
in vivo C57/BL6 mice bearing a subcutaneously transplanted B16 melanoma Si(IV)-methoxyethylene-glycol-naphthalocyanine
[154]
in vivo Syrian Golden hamsters fitted with dorsal skinfold chambers containing A-Mel-3 melanoma cells 5-aminolaevulinic acid (ALA) [155]
in vivo 10 choroidal melanomas in rabbits liposomal preparation of benzoporphyrin derivative [156]
in vivo M2R mouse melanoma tumors implanted in CD1 nude mice bacteriochlorophyll-serine (Bchl-Ser), [157]
in vitro uveal melanoma cells hematoporphyrin esters (HPE) [158]
in vivo Syrian Golden hamsters fitted with dorsal skinfold chambers containing A-Mel-3 melanoma cells 5-aminolaevulinic acid (ALA) [159]
in vivo C57/BL6 mice bearing a subcutaneously transplanted B16 melanoma Zn(II)-2,3 naphthalocyanine (ZnNc) [160]
in vivo 32 choroidal tumours in New Zealand albino rabbit eyes. benzoporphyrin derivative [161]
in vitro G361,M18 and M6 human melanoma cells hypericin [162]
in vitro melanoma cell lines hypericin [163]

Table 2.

Comprehensive update of in vivo and in vitro photodynamic therapy studies from 1996-present.

For melanoma treatment, where PDT will be more effective as a post-operative adjunctive treatment, very few reports highlight its effectiveness even though laboratory studies using melanoma cells show promise. Clinically, PDT has shown promise in the treatment of both ocular amelanotic melanomas [164] and skin metastases [165] however, more extensive clinical studies need to be conducted before PDT is accepted as the adjunctive therapy of choice [166] (Table 3).

Type of study Tumour Pigmentary phenotype Photosensitizer Outcome of study Ref
clinical choroidal melanoma unpigmented verteporfin Dramatic tumor regression over 2 months to a completely flat scar [1.3 mm thickness), and remained stable at 50 months of follow-up. [167]
clinical 3 choroidal melanomas pigmented PDT and intravitreal bevacizumab The tumors treated with PDT and bevacizumab showed a marked reduction in tumor vascularity. The tumors receiving PDT as a primary treatment were followed by progressive tumor growth that led to enucleation years after. [168]
clinical 9 posteriorly located choroidal melanomas unpigmented verteporfin Eight tumors demonstrated apparent complete regression over 1 month to 14 months with no recurrence during follow-up of between 34 months and 81 months. One case developed 2 separate local recurrences at 21 months and 34 months. [169]
clinical 11 late stage melanomas (cutaneous metastases) ND indocyanine green and imiquimod (immune modifier) Complete response was observed in 6 patients. All lesions in the treatment area of the patients responded to photo-immunotherapy, 8 of which achieved complete local response (CLR). CLR was observed in the non-treatment site (regional) lesions in four patients. Five patients were still alive at the time of last follow-up. [170]
clinical 1 duodenal metastatic melanoma ND porfirmer sodium Successful treatment. [171]
clinical 6 brain metastasis of malignant melanoma ND porfirmer sodium All 6 patients [100%) remained free of brain disease till death, 50% died of malignant melanoma elsewhere and 50% died of unrelated causes. [172]
clinical melanoma in situ unigmented methyl aminolevulinate (MAL) Recurrence at the original tumour site 4 months after PDT. [173]
clinical choroidal melanoma unpigmented benzoporphyrin derivative (BPD) The tumor fully disappeared 1 month after the treatment, the visual acuity improved from 4/16 to 4/4. The disease did not recur during 24-month follow-up. [174]
clinical 2 late-stage melanoma. Patient 1 had the primary tumour and local metastases on the left arm and metastatic tumours in the lungs. Patient 2 had a head and neck melanoma with multiple local metastases, which had failed repeated attempts at surgical resection and high-dose radiation therapy. pigmented indocyanine
green (ICG) + imiquimod (toll-like receptor
agonist)
Patient 1 free of all clinically detectable tumours (including the lung metastases) "/>20 months after the first treatment cycle. Patient 2 has been free of any clinical evidence of the tumour for over 6 months. [175]
clinical 4 uveal melanoma, PDT on actual tumour site mildly to heavily pigmented benzoporphyrin derivative (BPD) Vascular occlusion and thrombosis in mildly pigmented melanoma but no response in pigmented ones. [176]
clinical 25 small and medium choroidal melanomas ND indocyanine green and transpupillary thermotherapy After a mean of 2.4 treatments (range, 1 to 5 treatments), all of the tumors but one showed a significant volume reduction without clinical evidence of recurrences. Complications included retinal vascular occlusions, edema and superficial scarring of the macula, and rhegmatogenous retinal detachment. [177]
clinical 14 skin metastasis from malignant melanoma, despite multiple courses of chemotherapy pigmented chlorin e6 Complete regression after the first PDT treatment in eight cases and complete regression after multiple treatments in six cases. 11 of 14 patients died due to the progression of the melanoma, the median survival time after surgery was 883 days. [165]
clinical 38 choroidal melanomas ND indocyanine green (ICG) Changes in microcirculation 6 months after PDT, as well as significant decrease of tumors thickness in ultrasonography (mean 38%), were detected in all cases. Complete regression of intrinsic vessels was demonstrated by ICGA in 26 cases, and partial regression of pathological vascularization was found in 12 patients. [178]
clinical 4 choroidal melanomas ND benzoporphyrin derivative (BPD) One tumor decreased in size and remained stable for 18 months. One tumor had no growth for 11 months. Two melanomas continued to grow, necessitating enucleation. [179]
clinical 3 uveal melanoma, PDT on unaffected areas before enucleation ND benzoporphyrin derivative (BPD) Vascular thrombosis. No damage to the photoreceptors. [180]
clinical 36 uveal melanomas various hematoporphyrin derivative 76 % of tumours were not growing at the end of the first year, 62 % after the second year and 38% after the fifth year. No eyes were lost as a result of PDT. The degree of tumour pigmentation and patient age at therapy significantly influence the tumour response to PDT. [181]

Table 3.

Clinical reports and outcomes of photodynamic effectiveness of photodynamic therapy protocols including the melanoma pigmentary phenotype.

1.3.2. Hypericin, a second generation photosensitizer for PDT

Hypericin, a second generation PS isolated from the plant Hypericum perforatum, is a phenanthroperylenequinone with two broad peaks of absorption – 300-400nm (ultraviolet) and 500-600nm (white light) (Figure 1). This may be considered as a disadvantage as a number of current second-generation photosensitizers have absorption peaks beyond 630nm allowing for increased penetration into tissues [182]. However, white light, used to activate hypericin, does penetrate deep into the dermis of the skin. Moreover, activation with ultraviolet light could be a distinct advantage for the use of hypericin in daylight-mediated PDT. This type of PDT is more convenient for patients and clinicians and causes less pain. It poses a particularly interesting avenue to explore for hospitals in developing countries where space is limited and budgets are inadequate. Daylight-mediated PDT is an effective treatment for thin actinic keratosis, as shown in three randomized controlled clinical studies (reviewed in Wiegell et al., 2011) [183]. The potential of hypericin in clinical practice has been highlighted by reports on its use to treat squamous and basal cell carcinomas [184-187], pancreatic tumors [188], bladder carcinomas [189-193], nasophyrangeal tumors [194,195] and recently melanomas [196].

Figure 1.

Absorbance spectrum of hypericin. Box, the wavelength of light used in our studies [98,197] representing one of the two activation peaks. Inset, chemical structure of hypericin.

1.3.3. Melanoma cell death and biological mechanisms induced by hypericin-PDT

Despite these promising studies, very few reports have highlighted hypericin’s role in targeting melanoma. For the most part, cytotoxicity testing of new photosensitizers are tested on cell lines in vitro using assays such as the 3-[4,5-Dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide (MTT), which is a colorimetric assays for measuring the activity of enzymes that reduce MTT to formazan dyes, giving a purple color [198,199]. Other tests for cytotoxicity include dead cell protease tests. Despite being used as the “gold standard” for cytotoxicity testing, it must be borne in mind that these assays are based on cellular metabolic activity and could result in a false positive result were the treatment to produce a cytostatic effect in cells. Moreover, as these are colorimetric-based tests, the photosensitizer used itself may interfere with the wavelength at which these tests are read.

One of the first few reports testing 1 to 20mg/ml hypericin efficacy on squamous carcinoma, sarcoma and melanoma cell lines found that a combination of activating laser light sources resulted in a reduction in cell viability of 90% [163]. Following this, Hadjur et al. (1996) exposed human pigmented and unpigmented melanoma cell lines to hypericin and showed minimal cytotoxicity on uptake but upon activation with white light, increased cell death in all three cell lines. Their findings thus suggest that amelanotic melanomas may be more susceptible to hypericin-PDT than pigmented melanomas. Their possible reasons for this related to the presence of melanin and antioxidant status of melanomas [162].

Work from our laboratory has shown a pigmentation dependant susceptibility of melanoma cells to hypericin-PDT, with pigmented cells being less susceptible than unpigmented cells [54,98,200,201]. Upon depigmentation with tyrosinase inhibitors, kojic acid and phenylthiourea, pigmented melanoma cells become more susceptible to hypericin-PDT [54,69]. Moreover, 72 hours after hypericin-PDT the cell viability of the depigmented melanoma cells remained significantly less than the control cells. Over the same time period the cells not treated with kojic acid approached a cell viability similar to the control.

Melanin is a potent antioxidant which could be a reason for the increased resistance of pigmented melanoma cells to PDT due to the scavenging of ROS produced by this therapy. Indeed we have shown that after depigmenting melanoma cells with kojic acid more ROS is produced upon treatment with hypericin-PDT compared to pigmented melanoma cells which were not depigmented [54]. We did not find a difference between the caspase 3, 7 activity after hypericin-PDT for both the depigmented and pigmented melanoma cells, which was lower than control. This suggests that pigmented melanoma cells might induce a caspase-independent mode of cell death such as the activation of apoptosis-inducing factor (AIF). Moreover, these cells might also undergo necrosis, necroptosis or autophagy in response to hypericin-PDT. We have further shown induction of autophagy at 4hours after hypericin-PDT in both pigmented and unpigmented melanoma cells [197]. Interestingly, pigmented melanoma cells (UCT Mel-1) show higher levels of externalisation of Annexin V, an early apoptotic event, compared to mildy and unpigmented melanoma cells [501mel and A375, respectively) after hypericin-PDT (Figure 2). However, the cell death response of pigmented and unpigmented melanoma cells is very complex and does seem to be cell type dependant. A possible explanation for this may be that the cell lines used in our studies are from different genetic origins and they thus might differ in various biochemical characteristics, including their antioxidant systems. The subcellular localisation of the photosensitizer is another factor determining the cell death mode initiated by PDT. Upon activation by light, photosensitizers produce ROS which are short-lived species acting directly in their vicinity of production. Localisation to different cellular compartments thus induces different modes of cell death.

Note: Since the discovery of programmed cell death in the 1960’s the cell death field has evolved immensely. Researchers have shifted from morphological classifications to using more biochemical criteria. The increase in cell death studies necessitated a systemic classification of cell death modalities, which led to the formation of the Nomenclature Committee on Cell Death (NCCD). The main mission of this committee is ‘to provide a forum in which names describing distinct modalities of cell death are critically evaluated and recommendations on their definition and use are formulated, hoping that a non-rigid, yet uniform nomenclature will facilitate the communication among scientists and ultimately accelerate the pace of discovery’ [202-204].

Figure 2.

Graphs representing fluorescent activated cell sorting (FACS) analyses of melanoma cells at 30min, 1, 4, 7 and 24h after hypericin-PDT treatment (3µM hypericin with 1 J/cm2 UVA). A: unpigmented A375, B: mildly pigmented 501mel and C: pigmented UCT Mel-1. Cells were stained for early apoptosis (FITC Annexin V, BD Biosciences) and necrosis (LIVE/DEAD Fixable Violet stain, Invitrogen). Different modes of cell death are represented as proportional percentages normalised to the control, black: late apoptotic/ necrotic, dark grey: necrotic, light grey: apoptotic, white: live; n=3.

1.4. PDT targets to treat melanoma

1.4.1. Cell proliferation and survival

It is now well established that one of the chief characteristics of cancer cells is their ability to overcome cellular control of proliferation [205]. In melanocytes, proliferation is caused by a combination of several mitogenic growth factors such as stem cell factor (SCF), epidermal growth factor (EGF), fibroblast growth factor (FGF) and hepatocyte growth factor (HGF) which cause a sustained extracellular receptor kinase (ERK) activity [206]. In melanoma, the RAS/Raf/MEK/ERK pathway is a key regulating pathway in proliferation with ERK being hyperactivated in up to 90% of human melanomas [207]. BRAF and PTEN mutations (see above) are co-incident in about 20% of cases [208]. The most common mutation in BRAF is a glutamic acid for valine substitution at position 600 (V600EBRAF) [209]. This mutation leads to constitutive ERK signalling resulting in hyperproliferation and cell survival [210]. This pathway, through the EGF receptor as an extracellular ligand, has been a worthwhile target for PDT in that sustained activation of the ERK pathway protected cells from photofrin-based PDT as well as a reduction in the Raf protein levels in treated cells [211].

Nuclear factor kappa beta (NF-κβ) signalling leads to transcriptional regulation of a number of genes involved in responses ranging from proliferation, metastasis, and survival to inflammation. It therefore is an important target in PDT to stop aberrant cell proliferation. PDT-induced oxidative stress through increased ROS production has been shown to activate (NF-κβ) [212] and inactivate its inhibitor (Iκβ). Moreover, Ryter and Gomer showed increased NF-κβ binding in response to PDT stress in mouse cancer cells leading to a reduction in proliferation [213].

1.4.2. Inhibition of apoptosis

Apoptosis, a controlled mode of cell death, is characterised by cell shrinkage, chromatin condensation, DNA fragmentation, membrane blebbing and activation of caspases [214]. It is now well established that activation of the caspase cascade occurs through death receptor activation (extrinsic pathway) or through mitochondrial outer membrane permeabilization (intrinsic pathway). Both of these pathways have been shown to be activated through PDT. Several biochemical studies have established that PDT with different photosensitizers, including hypericin, utilise the mitochondrial-mediated pathway of caspase activation [215,216] although PDT has recently been shown to also engage caspase-independent pathways [217]. It is further known that several anti-cancer agents induce apoptosis and may share common pathways leading to cell killing with Fas/APO-1/CD95 [218,219]. Ali et al. (2002) elegantly showed that hypericin-PDT induces human nasopharyngeal cancer cells to undergo apoptosis through the Fas/FasL system. Moreover, they showed that the upregulation of Fas/FasL results in the release of cytochrome c into the cytoplasm with subsequent caspase induction – results that suggest that although apoptosis is considered a product of either an extrinsic or intrinsic mechanism; the overall response to PDT may be a combination of mechanisms [220].

Cancer cells are known to resist cell death by upregulation of anti-apoptotic proteins, mutations in pro-apoptotic proteins, inhibition of cell senescence or through protective mechanisms such as autophagy. PDT that is targeted for cancer therapy aims to invoke cell cytoxicity through attacking these characteristics – topics of a number of recent reviews [36,221-223].

Intriguingly, the emergence of a defined ‘immunogenic apoptosis’ seems to be a new ‘subset’ of apoptosis and autophagic cell death which has been shown to have the ability to release/expose damage-associated membrane proteins (DAMPs) [224-227]. Therapeutically, the immunogenicity of apoptosis is preferable for application rather than necrosis (or for that matter autophagic cell death) since necrosis can lead to harmful immunological reactions [228] (on the other hand, the extent of immunological impact of autophagic cell death is as yet uncharacterized, thereby making it an uncertain modality to use in the context of ‘immunochemotherapy’) [229]. The cell killing effectiveness is however dependent on parameters such as the PS used, the light dose and most importantly, the subcellular localization of the PS. It is crucial for photosensitizers to effectively enter the cell and accumulate in specific intracellular organelles in order to be efficient in their killing ability. Clearly, the final destination of the PS and its immediate vicinity will lead to different modes of cell death and consequently different efficiencies. Recent reports have highlighted that hypericin not only localises to different subcellular organelles but that this localization is exposure and dose-dependent in addition to being tumour cell-specific [230-234]. As a start however, the lipophillic nature of hypericin dictates its association with cellular membranes [235]. The fact that hypericin has been shown to associate with serum proteins (LDL and HDL lipoproteins [236] ensues that it enters cells quickly and is preferentially taken up by cancer cells in the 3-dimensional milieu as recent reports showed that these cells have high levels of LDL surface receptors [237]. This is further supported by a recent report showing that cholesterol serves as a key determinant for the uptake of hypericin into cellular membranes [238].

Noteworthy however is that even though high levels of hydrophobicity ensues, high levels of intracellular accumulation of the photosensitizer, changes in the physical structure of the PS due to aggregation and other modifications, may lead to reduced PDT efficiency [239]. Overall, the consensus emerging is that hypericin localises to three intracellular organelles namely, the endoplasmic reticulum (ER)-Golgi network [230,231,240,241], mitochondria (Mt) [242-245] and lysosomes [237,246] where through synergistic action, apoptosis is induced. More recent work by the Agostinis group show that hypericin-based PDT would produce photo-oxidative ER (p-ox ER stress) stress while 5-ALA (localizes in the mitochondria)-based PDT would produce photo-oxidative mitochondrial stress [36,247]. They also observed that Hyp-PDT induces ‘pre-apoptotic’ active exo-ATP secretion and late stage passive release of DAMPs like HSP70, HSP90 and CRT [223]. Overall they suggest that the potential of Hyp-PDT in causing exposure/secretion of ‘critical’ DAMPs add to the apoptotic cell death modality in a rather ‘small club’ of anti-cancerous therapeutic agents/modalities capable of exposing immunogenic signals like ecto-CRT [227,248].

1.4.3. Induction of autophagy

A recent finding is the induction of the cytoprotective programme of autophagy in melanomas in response to PDT-induced oxidative stress [89]. In addition, recent reports showed that cancer cells may respond to chemotherapeutics or other forms of oxidative stress such as PDT, through the induction of autophagy initially but continued stress leads to an overwhelming of the endogenous antioxidant enzymes along with a shift from autophagy to a possible senescent phenotype in an attempt to prolong cellular survival. Consequently however, the cell enters an apoptotic or necrotic mode of cell death [249-251]. Autophagy, defined as a cellular response to nutrient deprivation with consequent organelle breakdown, could converge with PDT at a number of cellular locations. Although more work relating to this aspect in melanomas is needed, reports on other cancer cells have shown that autophagy can be induced if the lysosomal system, needed for the clearance of ROS-damaged organelles, is affected by PDT [252]. Another cellular location is the mitochondria, where the PDT-induced loss of anti-apoptotic protein Bcl-2, may lead to an initation of autophagy [253].

1.4.4. Chemoresistance due to increased antioxidants

Cancer cells are considered to be under continuous oxidative stress which has been suggested to aid in tumor progression [254]. In support, several studies have shown tumor cell lines producing higher levels of ROS compared to their normal counterparts [255,256]. Due to this increased level of ROS and hence constitutive increased level of oxidative stress, it is not surprising that cancer cells have an extensive and advanced intracellular antioxidant network – a characteristic which further increases their chemoresistant property. Interestingly, the antioxidant status of melanomas differs from that of other skin cancers such as basal and squamous cell carcinomas in that their antioxidant activity levels (i.e. catalase, glutathione peroxidise, superoxide dismutase) are much higher [257]. In contrast, melanocytes, their normal untransformed phenotype, have lower levels of antioxidant activities and associated lower levels of resistance to oxidative stress [258]. It is therefore reasonable to postulate whether breaking this tolerance to oxidative stress may increase therapeutic efficacy in targeting melanoma. A number of studies have therefore suggested that treating melanoma by inhibiting cellular antioxidants may be efficacious [259-261]. One example of this was the addition of the superoxide dismutase (SOD) activity inhibitor, 2-methoxyestradiol (2-ME2), to a mouse transplant model which induced growth arrest of melanoma cells after injection [262]. Paradoxically, several studies have suggested that antioxidants can enhance the action of cancer chemotherapeutics drugs in their in vitro models through inhibition of a variety of factors which contribute to the malignant phenotype [263,264]. A recent study however, using six different combinations of antioxidants and chemotherapeutic drugs in combination, failed to identify a single combination in which an antioxidant reduced the survival of malignant breast carcinoma cells [265]. To our knowledge the use of PDT as an inhibitor of antioxidants has not been tested in cancer cells.

1.4.5. Melanin and melanosomes as pro-survival agents

All the potential intracellular organelle targets for PDT mentioned above are consistent with most cancer cells. However, the one aspect that sets melanoma apart from other cancers is the presence of its cell-specific organelle called the melanosome and its associated product, melanin pigment. It is thus not inconceivable to believe that the intractability of this skin disease may in some way be related to this organelle and its function [266,267]. It follows logically then, that treatment regimes need to consider the melanosome as another potential target organelle in the fight against melanoma [268].

Melanosomes are membrane-bound organelles in melanocytic cells which house the pathway that results in the formation of the polymeric pigment, melanin [269,270]. The enzymes which participate in this pathway are translated in the cytoplasm and chaperoned to the melanosomes. Tyrosinase (TYR), the rate-limiting enzyme of the pathway, and its related proteins tyrosinase-related proteins 1 and 2 (TYRP-1 and TYRP-2) act in concert to first convert tyrosine to 3,4-dihydroxy-phenylalanine (DOPA) via tyrosine hydroxylase activity and then convert DOPA to DOPAquinone via dopa oxidase activity. Both of these activities occur via separate tyrosinase catalytic sites. During melanin synthesis toxic intermediates such as 5, 6-dihydroxyindole and 5,6-dihydroxyindole-2-carboxylic acid are produced. Structurally, the melanosomes are designed to compartmentalise these cytotoxic melanin intermediates from spilling into the cytoplasm [271]. Melanosomal biogenesis progresses through four distinct stages of maturation where the first two stages contain no melanin and the later stages constitute intermediates required to generate a matrix favourable for the formation of melanin [269,272]. The Pmel17/gp100/Silv/ME20 protein, a product of the Silver locus in melanocytic cells [273], is capable of polymerizing into fibrillar arrays that form the backbone of melanosomes. As a major component of the fibrillar matrix of early stage melanosomes, Pmel-17 serves as the best marker to follow intracellular trafficking steps that regulate melanosomal formation [274].

Moreover, as Pmel-17 facilitates melanin deposition and plays a pivotal role in melanosome biogenesis, it remains a strategic target when trying to combat melanoma through the fact that melanosomes are involved in scavenging endogenous cytotoxic metabolites and storing their waste products - a function that has been suggested to be a key in creating multi-drug resistance [267]. With the premise that melanosomes may be acting as cytotoxic drug “sinks” through the sequestration of chemotherapeutic drugs [267], it would be considered an effective therapy for a PS to enter the melanosomal membrane and damage the wall of the melanosome thus allowing the leakage of toxic melanin intermediates resulting in cell death. The drawback is that melanosomes, which are classified into stages along their biogenesis [269,270] only produce the toxic intermediates during their final maturing stages III and IV [268-270,275-277]. Most pigmented melanomas do however present with a majority of these end-stage melanosomes in their cytoplasm making the melanosomal membrane an attractive target for PDT. On the basis of this information, one may imagine that pigmented melanomas are therefore more susceptible to PDT-induced cell death. In contrast, our work has shown that pigmented melanomas are much less susceptible to hypericin-PDT than unpigmented/amelanotic melanomas despite hypericin readily entering the melanosomes [89]. We hypothesize that the reason for this is due to the presence of the pigment melanin. In support of this, pigmented human xenograft melanotic melanoma in mice, was shown to be far less responsive to PDT than amelanotic melanoma [278].

Melanin has been shown to act as both an oxidant and antioxidant [266,279] and in parallel studies, its presence in melanomas have been linked to chemoresistance. In support, further studies have shown that a lack of pigment in melanomas decreases their resistance to cell death. Our ongoing investigation into susceptibility to PDT-induced cell death in depigmented melanomas supports this hypothesis [54,69].

1.4.6. Cancer stem cells as future PDT targets

The cancer stem cell hypothesis purports the idea that a subset of cancer cells is capable of maintaining and driving disease progression [280,281]. With the identification of cancer stem cell populations in colon, breast and brain tumors [282-285], it is believed that these cells are integrally related to tumor formation, resistance to chemotherapy and escape from remission [286]. While the qualifications for melanoma stem cells have generally been defined as tumorigenicity in xenograft spheroid formation and self-renewal in non-adherent cultures, the markers used to identify these cells from the general tumor population remain debatable. A brief summary of these markers and their potential as targets for novel PDT-based therapy, follow.

ATP-binding cassette (ABC) transporters are a vast family of transmembrane proteins that have been studied for their ability to actively transport cytotoxic substances out of cells [287]. Intriguingly, some of these transporters have been demonstrated to be highly expressed in highly tumorigenic subpopulations of melanoma suggesting that they may be markers of melanoma stem cells [286]. One of these includes the ABCB5 transporter. Known for increased expression during melanoma progression in human tumor samples, ABCB5+ cells were able to resist treatment with doxorubicin [288]. While ABCB5+ cells were not able to renew in culture (a “stemness trait”), a subpopulation of cells that were indeed able to renew expressed the ABC transporter, Multi-drug Resistant-1 (MDR1) [289]. In vitro, MDR1+ cells exhibited less pigmentation than MDR1- cells, possessed the ability to continuously self-renew in soft agar and expressed the pluripotency and self-renewal regulators, human telomerase reverse transcriptase (hTERT) – all characteristics pointing towards “stemness”. Interestingly, while MDR+ cells did exhibit cancer stem-cell like properties in vitro, they also co-expressed ABCB5 and ABCC2 mRNAs suggesting that a number of ABC transporters may be expressed in sub-populations [289]. To further add to the complication of delineating melanoma stem cell markers as potential targets for PDT is the fact that a number of recent markers are co-expressed with ABC transporters. These include CD133/prominin-1/AC133, which is co-expressed with ABCB5 and ABCG2 [288,290] and Nestin ([286].

Accumulating evidence suggests that another transporter, ABCG2, has physiological relevance in terms of photosensitivity and hence, PDT [291,292]. It has been shown that clinical photosensitizers and chemotherapeutic drugs have been transported out of cells by ABCG2 whereas this effect was abrogated by co-administration of its inhibitor, imatinib mesylate [293]. It is fascinating to speculate that a PDT protocol using a new, more stable photosensitizer such as hypericin may, through optimized concentrations, inhibit the action of the ABCG2 transporter and thus create an intracellular pool of ROS resulting in efficacious cell death. This is definitely an avenue for exploration.

Overall, the ability to halt melanoma cancer progression through targeting melanoma stem cells could be extremely advantageous. However, with such a large number of potential markers and their interaction with PDT unknown, it may be a better option to focus on ABC transporters and investigate their susceptibility to second generation PS-based PDT as a means to an end for melanoma progression.

Advertisement

2. Conclusion and future directions

There is no doubt that our understanding of the molecular and cellular basis of melanoma has grown substantially over the past decade. However, due to its multifunctional nature, the nee d for better, improved therapies to combat or target melanoma remain essential. In addition, better understanding of the heterogenous nature of this diverse disease will likely lead to re-evaluation of the basic concepts underlying melanoma therapeutics development and clinical trial design. Till then however, novel adjuvant treatment modalities such as PDT using photostable, second-generation photosensitizers such as hypericin remain an option and need to be investigated further. Moreover, optimization of this type of therapy with regard to subcellular localization and its effect on cell death mechanisms within melanoma cells is needed. Targeting the integrity of melanocytes-specific organelles such as the melanosomes and producing an over-riding increase in ROS with consequent cytotoxicity remains a good therapeutic option but needs a systematic, scientific approach. Intriguingly, as more avenues of therapeutic targets such as melanoma stem cells and ABC transporters become illuminated, the ability to invoke cell death modalities in combination with PDT become more evident. Finally, it is clear that all these factors need to be considered in synergy if progress is to be made toward combating the menace that is metastatic melanoma.

Advertisement

Acknowledgments

This work was supported by the University of Cape Town Research Committee, the National Research Foundation of South Africa (LMD) and Postgraduate funding (BK) from the NRF and DAAD.

References

  1. 1. Armstrong B. Epidemiology of cutaneous melanoma and current trends. In: Thompson J, Morton D, Kroon B, editors. Textbook of Melanoma London, UK: Martin Dunitz; 2004. p. 65-80.
  2. 2. Bataille V. Early detection of melanoma improves survival. Practitioner 2009 Oct;253(1722):29-32, 3.
  3. 3. Zbytek B, Carlson JA, Granese J, Ross J, Mihm MC, Slominski A. Current concepts of metastasis in melanoma. Expert Rev Dermatol 2008 Oct;3(5):569-585.
  4. 4. Carlson JA, Ross JS, Slominski A, Linette G, Mysliborski J, Hill J, et al. Molecular diagnostics in melanoma. J Am Acad Dermatol 2005 May;52(5):743-75; quiz 775-8.
  5. 5. Moncrieff MD, Martin R, O'Brien CJ, Shannon KF, Clark JR, Gao K, et al. Adjuvant postoperative radiotherapy to the cervical lymph nodes in cutaneous melanoma: is there any benefit for high-risk patients? Ann Surg Oncol 2008 Nov;15(11):3022-3027.
  6. 6. Yamamura H, Ugawa S, Ueda T, Morita A, Shimada S. TRPM8 activation suppresses cellular viability in human melanoma. Am J Physiol Cell Physiol 2008 Aug;295(2):C296-301.
  7. 7. Chudnovsky Y, Khavari PA, Adams AE. Melanoma genetics and the development of rational therapeutics. J Clin Invest 2005 Apr;115(4):813-824.
  8. 8. Slominski A, Tobin DJ, Shibahara S, Wortsman J. Melanin pigmentation in mammalian skin and its hormonal regulation. Physiol Rev 2004 Oct;84(4):1155-1228.
  9. 9. Slominski A, Wortsman J, Carlson AJ, Matsuoka LY, Balch CM, Mihm MC. Malignant melanoma. Arch Pathol Lab Med 2001 Oct;125(10):1295-1306.
  10. 10. Thompson JF, Scolyer RA, Kefford RF. Cutaneous melanoma. Lancet 2005 Feb 19-25;365(9460):687-701.
  11. 11. McMasters KM, Edwards MJ, Ross MI, Reintgen DS, Martin RC,2nd, Urist MM, et al. Ulceration as a predictive marker for response to adjuvant interferon therapy in melanoma. Ann Surg 2010 Sep;252(3):460-5; discussion 465-6.
  12. 12. Chapman PB, Hauschild A, Robert C, Haanen JB, Ascierto P, Larkin J, et al. Improved survival with vemurafenib in melanoma with BRAF V600E mutation. N Engl J Med 2011 Jun 30;364(26):2507-2516.
  13. 13. Wheatley K, Ives N, Hancock B, Gore M, Eggermont A, Suciu S. Does adjuvant interferon-alpha for high-risk melanoma provide a worthwhile benefit? A meta-analysis of the randomised trials. Cancer Treat Rev 2003 Aug;29(4):241-252.
  14. 14. Mocellin S, Pasquali S, Rossi CR, Nitti D. Interferon alpha adjuvant therapy in patients with high-risk melanoma: a systematic review and meta-analysis. J Natl Cancer Inst 2010 Apr 7;102(7):493-501.
  15. 15. Janku F, Kurzrock R. Adjuvant interferon in high-risk melanoma: end of the era? J Clin Oncol 2010 Jan 10;28(2):e15-6; author reply e17-8.
  16. 16. Boon T. Tumor antigens recognized by cytolytic T lymphocytes: present perspectives for specific immunotherapy. Int J Cancer 1993 May 8;54(2):177-180.
  17. 17. Vogel TU, Visan L, Ljutic B, Gajewska B, Caterini J, Salha D, et al. Preclinical qualification of a new multi-antigen candidate vaccine for metastatic melanoma. J Immunother 2010 Oct;33(8):743-758.
  18. 18. Gilchrest BA, Eller MS, Geller AC, Yaar M. The pathogenesis of melanoma induced by ultraviolet radiation. N Engl J Med 1999 Apr 29;340(17):1341-1348.
  19. 19. Satyamoorthy K, Herlyn M. Cellular and molecular biology of human melanoma. Cancer Biol Ther 2002 Jan-Feb;1(1):14-17.
  20. 20. Vogelstein B, Kinzler KW. Cancer genes and the pathways they control. Nat Med 2004 Aug;10(8):789-799.
  21. 21. Soengas MS, Lowe SW. Apoptosis and melanoma chemoresistance. Oncogene 2003 May 19;22(20):3138-3151.
  22. 22. Clark WH,Jr, Elder DE, Guerry D,4th, Epstein MN, Greene MH, Van Horn M. A study of tumor progression: the precursor lesions of superficial spreading and nodular melanoma. Hum Pathol 1984 Dec;15(12):1147-1165.
  23. 23. Brunet A, Roux D, Lenormand P, Dowd S, Keyse S, Pouyssegur J. Nuclear translocation of p42/p44 mitogen-activated protein kinase is required for growth factor-induced gene expression and cell cycle entry. EMBO J 1999 Feb 1;18(3):664-674.
  24. 24. Lin AW, Barradas M, Stone JC, van Aelst L, Serrano M, Lowe SW. Premature senescence involving p53 and p16 is activated in response to constitutive MEK/MAPK mitogenic signaling. Genes Dev 1998 Oct 1;12(19):3008-3019.
  25. 25. Miller AJ, Mihm MC,Jr. Melanoma. N Engl J Med 2006 Jul 6;355(1):51-65.
  26. 26. Kefford R, Bishop JN, Tucker M, Bressac-de Paillerets B, Bianchi-Scarra G, Bergman W, et al. Genetic testing for melanoma. Lancet Oncol 2002 Nov;3(11):653-654.
  27. 27. Serrano M. The tumor suppressor protein p16INK4a. Exp Cell Res 1997 Nov 25;237(1):7-13.
  28. 28. Gilchrest BA, Eller MS. DNA photodamage stimulates melanogenesis and other photoprotective responses. J Investig Dermatol Symp Proc 1999 Sep;4(1):35-40.
  29. 29. Johnson TM, Dolan OM, Hamilton TA, Lu MC, Swanson NA, Lowe L. Clinical and histologic trends of melanoma. J Am Acad Dermatol 1998 May;38(5 Pt 1):681-686.
  30. 30. Rossi CR, Foletto M, Vecchiato A, Alessio S, Menin N, Lise M. Management of cutaneous melanoma M0: state of the art and trends. Eur J Cancer 1997 Dec;33(14):2302-2312.
  31. 31. Slominski A, Ross J, Mihm MC. Cutaneous melanoma: pathology, relevant prognostic indicators and progression. Br Med Bull 1995 Jul;51(3):548-569.
  32. 32. Clark WH,Jr, Elder DE, Guerry D,4th, Braitman LE, Trock BJ, Schultz D, et al. Model predicting survival in stage I melanoma based on tumor progression. J Natl Cancer Inst 1989 Dec 20;81(24):1893-1904.
  33. 33. Guerry D,4th, Synnestvedt M, Elder DE, Schultz D. Lessons from tumor progression: the invasive radial growth phase of melanoma is common, incapable of metastasis, and indolent. J Invest Dermatol 1993 Mar;100(3):342S-345S.
  34. 34. Gray-Schopfer V, Wellbrock C, Marais R. Melanoma biology and new targeted therapy. Nature 2007 Feb 22;445(7130):851-857.
  35. 35. Jean D, Gershenwald JE, Huang S, Luca M, Hudson MJ, Tainsky MA, et al. Loss of AP-2 results in up-regulation of MCAM/MUC18 and an increase in tumor growth and metastasis of human melanoma cells. J Biol Chem 1998 Jun 26;273(26):16501-16508.
  36. 36. Buytaert E, Dewaele M, Agostinis P. Molecular effectors of multiple cell death pathways initiated by photodynamic therapy. Biochim Biophys Acta 2007 Sep;1776(1):86-107.
  37. 37. Babilas P, Schreml S, Landthaler M, Szeimies RM. Photodynamic therapy in dermatology: state-of-the-art. Photodermatol Photoimmunol Photomed 2010 Jun;26(3):118-132.
  38. 38. Dougherty TJ, Gomer CJ, Henderson BW, Jori G, Kessel D, Korbelik M, et al. Photodynamic therapy. J Natl Cancer Inst 1998 Jun 17;90(12):889-905.
  39. 39. Henderson BW, Dougherty TJ. How does photodynamic therapy work? Photochem Photobiol 1992 Jan;55(1):145-157.
  40. 40. Juarranz A, Jaen P, Sanz-Rodriguez F, Cuevas J, Gonzalez S. Photodynamic therapy of cancer. Basic principles and applications. Clin Transl Oncol 2008 Mar;10(3):148-154.
  41. 41. Ortel B, Shea CR, Calzavara-Pinton P. Molecular mechanisms of photodynamic therapy. Front Biosci 2009 Jan 1;14:4157-4172.
  42. 42. Gomer CJ, Ferrario A, Luna M, Rucker N, Wong S. Photodynamic therapy: combined modality approaches targeting the tumor microenvironment. Lasers Surg Med 2006 Jun;38(5):516-521.
  43. 43. Verma S, Watt GM, Mai Z, Hasan T. Strategies for enhanced photodynamic therapy effects. Photochem Photobiol 2007 Sep-Oct;83(5):996-1005.
  44. 44. Detty MR, Gibson SL, Wagner SJ. Current clinical and preclinical photosensitizers for use in photodynamic therapy. J Med Chem 2004 Jul 29;47(16):3897-3915.
  45. 45. Stockert JC, Canete M, Juarranz A, Villanueva A, Horobin RW, Borrell JI, et al. Porphycenes: facts and prospects in photodynamic therapy of cancer. Curr Med Chem 2007;14(9):997-1026.
  46. 46. Bourre L, Rousset N, Thibaut S, Eleouet S, Lajat Y, Patrice T. PDT effects of m-THPC and ALA, phototoxicity and apoptosis. Apoptosis 2002 Jun;7(3):221-230.
  47. 47. Thibaut S, Bourre L, Hernot D, Rousset N, Lajat Y, Patrice T. Effects of BAPTA-AM, Forskolin, DSF and Z.VAD.fmk on PDT-induced apoptosis and m-THPC phototoxicity on B16 cells. Apoptosis 2002 Apr;7(2):99-106.
  48. 48. Szeimies RM, Karrer S, Radakovic-Fijan S, Tanew A, Calzavara-Pinton PG, Zane C, et al. Photodynamic therapy using topical methyl 5-aminolevulinate compared with cryotherapy for actinic keratosis: A prospective, randomized study. J Am Acad Dermatol 2002 Aug;47(2):258-262.
  49. 49. Szeimies RM. Methyl aminolevulinate-photodynamic therapy for basal cell carcinoma. Dermatol Clin 2007 Jan;25(1):89-94.
  50. 50. Szeimies RM, Morton CA, Sidoroff A, Braathen LR. Photodynamic therapy for non-melanoma skin cancer. Acta Derm Venereol 2005;85(6):483-490.
  51. 51. Bolfarini GC, Siqueira-Moura MP, Demets GJ. In vitro evaluation of combined hyperthermia and photodynamic effects using magnetoliposomes loaded with cucurbituril zinc phthalocyanine complex on melanoma. 2012 Jun 5.
  52. 52. Khurana M, Ulrich S, Kim A, Moriyama Y, Netchev G, Akens MK, et al. Biodistribution and Pharmacokinetic Studies of a Porphyrin Dimer Photosensitizer (Oxdime) by Fluorescence Imaging and Spectroscopy in Mice Bearing Xenograft Tumors. Photochem Photo 2012 Jun 23.
  53. 53. Rapozzi V, Zorzet S, Zacchigna M, Drioli S, Xodo LE. The PDT activity of free and pegylated pheophorbide a against an amelanotic melanoma transplanted in C57/BL6 mice. Invest New Drugs 2012 Jun 12.
  54. 54. Sharma KV, Davids LM. Depigmentation in melanomas increases the efficacy of hypericin-mediated photodynamic-induced cell death. Photodiagnosis Photodyn Ther 2012 Jun;9(2):156-163.
  55. 55. Shrestha TB, Seo GM, Basel MT, Kalita M, Wang H, Villanueva D, et al. Stem cell-based photodynamic therapy. Photochem Photobiol Sci 2012 Jul;11(7):1251-1258.
  56. 56. Conforti F, Menichini G, Zanfini L, Tundis R, Statti GA, Provenzano E, et al. Evaluation of phototoxic potential of aerial components of the fig tree against human melanoma. Cell Prolif 2012 Jun;45(3):279-285.
  57. 57. Maduray K, Odhav B, Nyokong T. In vitro photodynamic effect of aluminum tetrasulfophthalocyanines on melanoma skin cancer and healthy normal skin cells. Photodiagnosis Photodyn Ther 2012 Mar;9(1):32-39.
  58. 58. Slastnikova TA, Rosenkranz AA, Gulak PV, Schiffelers RM, Lupanova TN, Khramtsov YV, et al. Modular nanotransporters: a multipurpose in vivo working platform for targeted drug delivery. Int J Nanomedicine 2012;7:467-482.
  59. 59. Wagner M, Suarez ER, Theodoro TR, Machado Filho CD, Gama MF, Tardivo JP, et al. Methylene blue photodynamic therapy in malignant melanoma decreases expression of proliferating cell nuclear antigen and heparanases. Clin Exp Dermatol 2012 Jul;37(5):527-533.
  60. 60. Dabrowski JM, Arnaut LG, Pereira MM, Urbanska K, Simoes S, Stochel G, et al. Combined effects of singlet oxygen and hydroxyl radical in photodynamic therapy with photostable bacteriochlorins: evidence from intracellular fluorescence and increased photodynamic efficacy in vitro. Free Radic Biol Med 2012 Apr 1;52(7):1188-1200.
  61. 61. Yoshii H, Yoshii Y, Asai T, Furukawa T, Takaichi S, Fujibayashi Y. Photo-excitation of carotenoids causes cytotoxicity via singlet oxygen production. Biochem Biophys Res Commun 2012 Jan 6;417(1):640-645.
  62. 62. Schmitt F, Barry NP, Juillerat-Jeanneret L, Therrien B. Efficient photodynamic therapy of cancer using chemotherapeutic porphyrin-ruthenium metalla-cubes. Bioorg Med Chem Lett 2012 Jan 1;22(1):178-180.
  63. 63. Menichini G, Alfano C, Provenzano E, Marrelli M, Statti GA, Menichini F, et al. Cachrys pungens Jan inhibits human melanoma cell proliferation through photo-induced cytotoxic activity. Cell Prolif 2012 Feb;45(1):39-47.
  64. 64. Radzi R, Osaki T, Tsuka T, Imagawa T, Minami S, Okamoto Y. Morphological study in B16F10 murine melanoma cells after photodynamic hyperthermal therapy with indocyanine green (ICG). J Vet Med Sci 2012 Apr;74(4):465-472.
  65. 65. Rapozzi V, Umezawa K, Xodo LE. Role of NF-kappaB/Snail/RKIP loop in the response of tumor cells to photodynamic therapy. Lasers Surg Med 2011 Sep;43(7):575-585.
  66. 66. Dabrowski JM, Krzykawska M, Arnaut LG, Pereira MM, Monteiro CJ, Simoes S, et al. Tissue uptake study and photodynamic therapy of melanoma-bearing mice with a nontoxic, effective chlorin. ChemMedChem 2011 Sep 5;6(9):1715-1726.
  67. 67. Kim MR, Morrison H, Mohammed SI. Effect of a photoactivated rhodium complex in melanoma. Anticancer Drugs 2011 Oct;22(9):896-904.
  68. 68. You BJ, Wu YC, Wu CY, Bao BY, Chen MY, Chang YH, et al. Proteomics displays cytoskeletal proteins and chaperones involvement in Hedyotis corymbosa-induced photokilling in skin cancer cells. Exp Dermatol 2011 Aug;20(8):653-658.
  69. 69. Sharma KV, Bowers N, Davids LM. Photodynamic therapy-induced killing is enhanced in depigmented metastatic melanoma cells. Cell Biol Int 2011 Sep;35(9):939-944.
  70. 70. Maduray K, Karsten A, Odhav B, Nyokong T. In vitro toxicity testing of zinc tetrasulfophthalocyanines in fibroblast and keratinocyte cells for the treatment of melanoma cancer by photodynamic therapy. J Photochem Photobiol B 2011 May 3;103(2):98-104.
  71. 71. Grimm S, Mvondo D, Grune T, Breusing N. The outcome of 5-ALA-mediated photodynamic treatment in melanoma cells is influenced by vitamin C and heme oxygenase-1. Biofactors 2011 Jan;37(1):17-24.
  72. 72. Tammela T, Saaristo A, Holopainen T, Yla-Herttuala S, Andersson LC, Virolainen S, et al. Photodynamic ablation of lymphatic vessels and intralymphatic cancer cells prevents metastasis. Sci Transl Med 2011 Feb 9;3(69):69ra11.
  73. 73. Kastle M, Grimm S, Nagel R, Breusing N, Grune T. Combination of PDT and inhibitor treatment affects melanoma cells and spares keratinocytes. Free Radic Biol Med 2011 Jan 15;50(2):305-312.
  74. 74. Robertson CA, Abrahamse H, Evans D. The in vitro PDT efficacy of a novel metallophthalocyanine (MPc) derivative and established 5-ALA photosensitizing dyes against human metastatic melanoma cells. Lasers Surg Med 2010 Dec;42(10):766-776.
  75. 75. Moisenovich MM, Ol'shevskaya VA, Rokitskaya TI, Ramonova AA, Nikitina RG, Savchenko AN, et al. Novel photosensitizers trigger rapid death of malignant human cells and rodent tumor transplants via lipid photodamage and membrane permeabilization. PLoS One 2010 Sep 15;5(9):e12717.
  76. 76. Otake E, Sakuma S, Torii K, Maeda A, Ohi H, Yano S, et al. Effect and mechanism of a new photodynamic therapy with glycoconjugated fullerene. Photochem Photobiol 2010 Nov-Dec;86(6):1356-1363.
  77. 77. Dabrowski JM, Arnaut LG, Pereira MM, Monteiro CJ, Urbanska K, Simoes S, et al. New halogenated water-soluble chlorin and bacteriochlorin as photostable PDT sensitizers: synthesis, spectroscopy, photophysics, and in vitro photosensitizing efficacy. ChemMedChem 2010 Oct 4;5(10):1770-1780.
  78. 78. Chen P, Jiang X, Wei DP, Li L. The effects of intense pulsed light and 5-aminolevulinic acid on the cultured B16 marine melanoma cells. Sichuan Da Xue Xue Bao Yi Xue Ban 2010 Mar;41(2):280-283.
  79. 79. Mroz P, Huang YY, Szokalska A, Zhiyentayev T, Janjua S, Nifli AP, et al. Stable synthetic bacteriochlorins overcome the resistance of melanoma to photodynamic therapy. FASEB J 2010 Sep;24(9):3160-3170.
  80. 80. Camerin M, Magaraggia M, Soncin M, Jori G, Moreno M, Chambrier I, et al. The in vivo efficacy of phthalocyanine-nanoparticle conjugates for the photodynamic therapy of amelanotic melanoma. Eur J Cancer 2010 Jul;46(10):1910-1918.
  81. 81. Barbugli PA, Siqueira-Moura MP, Espreafico EM, Tedesco AC. In vitro phototoxicity of liposomes and nanocapsules containing chloroaluminum phthalocyanine on human melanoma cell line. J Nanosci Nanotechnol 2010 Jan;10(1):569-573.
  82. 82. Serra A, Pineiro M, Santos CI, Gonsalves AM, Abrantes M, Laranjo M, et al. In vitro photodynamic activity of 5,15-bis(3-hydroxyphenyl)porphyrin and its halogenated derivatives against cancer cells. Photochem Photobiol 2010 Jan-Feb;86(1):206-212.
  83. 83. Nikitina RG, Mardynskaja VP, Morozova TG, Drozhzhina VV. A method for improving the efficiency of therapy for melanoma. Bull Exp Biol Med 2009 Aug;148(2):238-240.
  84. 84. Ouedraogo M, De Maesschalck E, Soentjens-Werts V, Dubois J. In vitro cytotoxicity study of oxaziridines generated after chlordiazepoxide, demoxepam, and desmethylchlordiazepoxide UV irradiation. Drug Chem Toxicol 2009;32(4):417-423.
  85. 85. Camerin M, Rello-Varona S, Villanueva A, Rodgers MA, Jori G. Metallo-naphthalocyanines as photothermal sensitisers for experimental tumours: in vitro and in vivo studies. Lasers Surg Med 2009 Nov;41(9):665-673.
  86. 86. Krestyn E, Kolarova H, Bajgar R, Tomankova K. Photodynamic properties of ZnTPPS(4), ClAlPcS(2) and ALA in human melanoma G361 cells. Toxicol In Vitro 2009 Aug 29.
  87. 87. Zhao B, Yin JJ, Bilski PJ, Chignell CF, Roberts JE, He YY. Enhanced photodynamic efficacy towards melanoma cells by encapsulation of Pc4 in silica nanoparticles. Toxicol Appl Pharmacol 2009 Dec 1;241(2):163-172.
  88. 88. Mamoon AM, Gamal-Eldeen AM, Ruppel ME, Smith RJ, Tsang T, Miller LM. In vitro efficiency and mechanistic role of indocyanine green as photodynamic therapy agent for human melanoma. Photodiagnosis Photodyn Ther 2009 Jun;6(2):105-116.
  89. 89. Davids LM, Kleemann B, Cooper S, Kidson SH. Melanomas display increased cytoprotection to hypericin-mediated cytotoxicity through the induction of autophagy. Cell Biol Int 2009 Oct;33(10):1065-1072.
  90. 90. Kolarova H, Tomankova K, Bajgar R, Kolar P, Kubinek R. Photodynamic and sonodynamic treatment by phthalocyanine on cancer cell lines. Ultrasound Med Biol 2009 Aug;35(8):1397-1404.
  91. 91. Tsai T, Ji HT, Chiang PC, Chou RH, Chang WS, Chen CT. ALA-PDT results in phenotypic changes and decreased cellular invasion in surviving cancer cells. Lasers Surg Med 2009 Apr;41(4):305-315.
  92. 92. Chen Y, Zheng W, Li Y, Zhong J, Ji J, Shen P. Apoptosis induced by methylene-blue-mediated photodynamic therapy in melanomas and the involvement of mitochondrial dysfunction revealed by proteomics. Cancer Sci 2008 Oct;99(10):2019-2027.
  93. 93. Hao E, Friso E, Miotto G, Jori G, Soncin M, Fabris C, et al. Synthesis and biological investigations of tetrakis(p-carboranylthio-tetrafluorophenyl)chlorin (TPFC). Org Biomol Chem 2008 Oct 21;6(20):3732-3740.
  94. 94. Chang CJ, Yu JS, Wei FC. In vitro and in vivo photosensitizing applications of Photofrin in malignant melanoma cells. Chang Gung Med J 2008 May-Jun;31(3):260-267.
  95. 95. Korbelik M, Cecic I. Complement activation cascade and its regulation: relevance for the response of solid tumors to photodynamic therapy. J Photochem Photobiol B 2008 Oct 16;93(1):53-59.
  96. 96. Kolarova H, Nevrelova P, Tomankova K, Kolar P, Bajgar R, Mosinger J. Production of reactive oxygen species after photodynamic therapy by porphyrin sensitizers. Gen Physiol Biophys 2008 Jun;27(2):101-105.
  97. 97. Skidan I, Dholakia P, Torchilin V. Photodynamic therapy of experimental B-16 melanoma in mice with tumor-targeted 5,10,15,20-tetraphenylporphin-loaded PEG-PE micelles. J Drug Target 2008 Jul;16(6):486-493.
  98. 98. Davids LM, Kleemann B, Kacerovska D, Pizinger K, Kidson SH. Hypericin phototoxicity induces different modes of cell death in melanoma and human skin cells. J Photochem Photobiol B 2008 May 29;91(2-3):67-76.
  99. 99. Schmitt F, Govindaswamy P, Suss-Fink G, Ang WH, Dyson PJ, Juillerat-Jeanneret L, et al. Ruthenium porphyrin compounds for photodynamic therapy of cancer. J Med Chem 2008 Mar 27;51(6):1811-1816.
  100. 100. Ma LW, Nielsen KP, Iani V, Moan J. A new method for photodynamic therapy of melanotic melanoma -- effects of depigmentation with violet light photodynamic therapy. J Environ Pathol Toxicol Oncol 2007;26(3):165-172.
  101. 101. Janczyk A, Wolnicka-Glubisz A, Urbanska K, Kisch H, Stochel G, Macyk W. Photodynamic activity of platinum(IV) chloride surface-modified TiO2 irradiated with visible light. Free Radic Biol Med 2008 Mar 15;44(6):1120-1130.
  102. 102. Tomankova K, Kolarova H, Vujtek M, Bajgar R. Photodynamic effect on melanoma cells investigated by atomic force microscopy. Gen Physiol Biophys 2007 Sep;26(3):200-206.
  103. 103. Frank J, Lornejad-Schafer MR, Schoffl H, Flaccus A, Lambert C, Biesalski HK. Inhibition of heme oxygenase-1 increases responsiveness of melanoma cells to ALA-based photodynamic therapy. Int J Oncol 2007 Dec;31(6):1539-1545.
  104. 104. Fabris C, Vicente MG, Hao E, Friso E, Borsetto L, Jori G, et al. Tumour-localizing and -photosensitising properties of meso-tetra(4-nido-carboranylphenyl)porphyrin (H2TCP). J Photochem Photobiol B 2007 Dec 14;89(2-3):131-138.
  105. 105. Rijcken CJ, Hofman JW, van Zeeland F, Hennink WE, van Nostrum CF. Photosensitiser-loaded biodegradable polymeric micelles: preparation, characterisation and in vitro PDT efficacy. J Control Release 2007 Dec 20;124(3):144-153.
  106. 106. Kolarova H, Nevrelova P, Bajgar R, Jirova D, Kejlova K, Strnad M. In vitro photodynamic therapy on melanoma cell lines with phthalocyanine. Toxicol In Vitro 2007 Mar;21(2):249-253.
  107. 107. Dabrowski JM, Pereira MM, Arnaut LG, Monteiro CJ, Peixoto AF, Karocki A, et al. Synthesis, photophysical studies and anticancer activity of a new halogenated water-soluble porphyrin. Photochem Photobiol 2007 Jul-Aug;83(4):897-903.
  108. 108. Ji HX, Zhang F, Gao LQ, Jia LY, Xiong Y, Liang C, et al. Photodynamic therapy of pigmented choroidal melanomas in rabbits using hematoporphyrin monomethyl ether. Zhonghua Yan Ke Za Zhi 2007 Mar;43(3):212-216.
  109. 109. Kolarova H, Lenobel R, Kolar P, Strnad M. Sensitivity of different cell lines to phototoxic effect of disulfonated chloroaluminium phthalocyanine. Toxicol In Vitro 2007 Oct;21(7):1304-1306.
  110. 110. Ferrario A, Rucker N, Wong S, Luna M, Gomer CJ. Survivin, a member of the inhibitor of apoptosis family, is induced by photodynamic therapy and is a target for improving treatment response. Cancer Res 2007 May 15;67(10):4989-4995.
  111. 111. Korbelik M, Cooper PD. Potentiation of photodynamic therapy of cancer by complement: the effect of gamma-inulin. Br J Cancer 2007 Jan 15;96(1):67-72.
  112. 112. Hiruma H, Katakura T, Takenami T, Igawa S, Kanoh M, Fujimura T, et al. Vesicle disruption, plasma membrane bleb formation, and acute cell death caused by illumination with blue light in acridine orange-loaded malignant melanoma cells. J Photochem Photobiol B 2007 Jan 3;86(1):1-8.
  113. 113. Nowak-Sliwinska P, Karocki A, Elas M, Pawlak A, Stochel G, Urbanska K. Verteporfin, photofrin II, and merocyanine 540 as PDT photosensitizers against melanoma cells. Biochem Biophys Res Commun 2006 Oct 20;349(2):549-555.
  114. 114. Schacht V, Szeimies RM, Abels C. Photodynamic therapy with 5-aminolevulinic acid induces distinct microcirculatory effects following systemic or topical application. Photochem Photobiol Sci 2006 May;5(5):452-458.
  115. 115. Saji H, Song W, Furumoto K, Kato H, Engleman EG. Systemic antitumor effect of intratumoral injection of dendritic cells in combination with local photodynamic therapy. Clin Cancer Res 2006 Apr 15;12(8):2568-2574.
  116. 116. Friso E, Roncucci G, Dei D, Soncin M, Fabris C, Chiti G, et al. A novel 10B-enriched carboranyl-containing phthalocyanine as a radio- and photo-sensitising agent for boron neutron capture therapy and photodynamic therapy of tumours: in vitro and in vivo studies. Photochem Photobiol Sci 2006 Jan;5(1):39-50.
  117. 117. Saczko J, Kulbacka J, Chwilkowska A, Drag-Zalesiniska M, Wysocka T, Lugowski M, et al. The influence of photodynamic therapy on apoptosis in human melanoma cell line. Folia Histochem Cytobiol 2005;43(3):129-132.
  118. 118. Kolarova H, Macecek J, Nevrelova P, Huf M, Tomecka M, Bajgar R, et al. Photodynamic therapy with zinc-tetra(p-sulfophenyl)porphyrin bound to cyclodextrin induces single strand breaks of cellular DNA in G361 melanoma cells. Toxicol In Vitro 2005 Oct;19(7):971-974.
  119. 119. Camerin M, Rello S, Villanueva A, Ping X, Kenney ME, Rodgers MA, et al. Photothermal sensitisation as a novel therapeutic approach for tumours: studies at the cellular and animal level. Eur J Cancer 2005 May;41(8):1203-1212.
  120. 120. Macecek J, Kolarova H, Psotova J, Bajgar R, Huf M, Nevrelova P, et al. Assessment of cellular damage by comet assay after photodynamic therapy in vitro. Acta Medica (Hradec Kralove) 2004;47(4):327-329.
  121. 121. Posen Y, Kalchenko V, Seger R, Brandis A, Scherz A, Salomon Y. Manipulation of redox signaling in mammalian cells enabled by controlled photogeneration of reactive oxygen species. J Cell Sci 2005 May 1;118(Pt 9):1957-1969.
  122. 122. Schastak S, Jean B, Handzel R, Kostenich G, Hermann R, Sack U, et al. Improved pharmacokinetics, biodistribution and necrosis in vivo using a new near infra-red photosensitizer: tetrahydroporphyrin tetratosylat. J Photochem Photobiol B 2005 Mar 1;78(3):203-213.
  123. 123. Mazor O, Brandis A, Plaks V, Neumark E, Rosenbach-Belkin V, Salomon Y, et al. WST11, a novel water-soluble bacteriochlorophyll derivative; cellular uptake, pharmacokinetics, biodistribution and vascular-targeted photodynamic activity using melanoma tumors as a model. Photochem Photobiol 2005 Mar-Apr;81(2):342-351.
  124. 124. Vena FC, Turchiello RF, Laville I, Pigaglio S, Blais J, Tedesco AC. 5-aminolevulinic acid ester-induced protoporphyrin IX in a murine melanoma cell line. Lasers Med Sci 2004;19(2):119-126.
  125. 125. Karmakova T, Feofanov A, Nazarova A, Grichine A, Yakubovskaya R, Luk'yanets E, et al. Distribution of metal-free sulfonated phthalocyanine in subcutaneously transplanted murine tumors. J Photochem Photobiol B 2004 Jul 19;75(1-2):81-87.
  126. 126. Ropp S, Guy J, Berl V, Bischoff P, Lepoittevin JP. Synthesis and photocytotoxic activity of new alpha-methylene-gamma-butyrolactone-psoralen heterodimers. Bioorg Med Chem 2004 Jul 1;12(13):3619-3625.
  127. 127. Ickowicz Schwartz D, Gozlan Y, Greenbaum L, Babushkina T, Katcoff DJ, Malik Z. Differentiation-dependent photodynamic therapy regulated by porphobilinogen deaminase in B16 melanoma. Br J Cancer 2004 May 4;90(9):1833-1841.
  128. 128. Lim DS, Ko SH, Lee WY. Silkworm-pheophorbide alpha mediated photodynamic therapy against B16F10 pigmented melanoma. J Photochem Photobiol B 2004 Mar 19;74(1):1-6.
  129. 129. Barge J, Decreau R, Julliard M, Hubaud JC, Sabatier AS, Grob JJ, et al. Killing efficacy of a new silicon phthalocyanine in human melanoma cells treated with photodynamic therapy by early activation of mitochondrion-mediated apoptosis. Exp Dermatol 2004 Jan;13(1):33-44.
  130. 130. Szurko A, Kramer-Marek G, Widel M, Ratuszna A, Habdas J, Kus P. Photodynamic effects of two water soluble porphyrins evaluated on human malignant melanoma cells in vitro. Acta Biochim Pol 2003;50(4):1165-1174.
  131. 131. Kolarova H, Mosinger J, Lenobel R, Kejlova K, Jirova D, Strnad M. In vitro toxicity testing of supramolecular sensitizers for photodynamic therapy. Toxicol In Vitro 2003 Oct-Dec;17(5-6):775-778.
  132. 132. Nagata S, Obana A, Gohto Y, Nakajima S. Necrotic and apoptotic cell death of human malignant melanoma cells following photodynamic therapy using an amphiphilic photosensitizer, ATX-S10(Na). Lasers Surg Med 2003;33(1):64-70.
  133. 133. Babilas P, Schacht V, Liebsch G, Wolfbeis OS, Landthaler M, Szeimies RM, et al. Effects of light fractionation and different fluence rates on photodynamic therapy with 5-aminolaevulinic acid in vivo. Br J Cancer 2003 May 6;88(9):1462-1469.
  134. 134. Haddad R, Kaplan O, Greenberg R, Siegal A, Skornick Y, Kashtan H. Photodynamic therapy of murine colon cancer and melanoma using systemic aminolevulinic acid as a photosensitizer. Int J Surg Investig 2000;2(3):171-178.
  135. 135. Jezek P, Nekvasil M, Skobisova E, Urbankova E, Jirsa M, Zadinova M, et al. Experimental photodynamic therapy with MESO-tetrakisphenylporphyrin (TPP) in liposomes leads to disintegration of human amelanotic melanoma implanted to nude mice. Int J Cancer 2003 Feb 20;103(5):693-702.
  136. 136. Hu L, Wu X, Song Y, Young LH, Gragoudas ES. Photodynamic therapy of pigmented choroidal melanomas in rabbits. Zhonghua Yan Ke Za Zhi 2002 Aug;38(8):491-494.
  137. 137. Urbanska K, Romanowska-Dixon B, Matuszak Z, Oszajca J, Nowak-Sliwinsk P, Stochel G. Indocyanine green as a prospective sensitizer for photodynamic therapy of melanomas. Acta Biochim Pol 2002;49(2):387-391.
  138. 138. Bourre L, Rousset N, Thibaut S, Eleouet S, Lajat Y, Patrice T. PDT effects of m-THPC and ALA, phototoxicity and apoptosis. Apoptosis 2002 Jun;7(3):221-230.
  139. 139. Lang K, Bolsen K, Stahl W, Ruzicka T, Sies H, Lehmann P, et al. The 5-aminolevulinic acid-induced porphyrin biosynthesis in benign and malignant cells of the skin. J Photochem Photobiol B 2001 Dec 1;65(1):29-34.
  140. 140. Drzewiecka A, Urbanska K, Matuszak Z, Pineiro M, Arnaut LG, Habdas J, et al. Tritolylporphyrin dimer as a new potent hydrophobic sensitizer for photodynamic therapy of melanoma. Acta Biochim Pol 2001;48(1):277-282.
  141. 141. Pahernik S, Langer S, Botzlar A, Dellian M, Goetz AE. Tissue distribution and penetration of 5-ALA induced fluorescence in an amelanotic melanoma after topical application. Anticancer Res 2001 Jan-Feb;21(1A):59-63.
  142. 142. Zilberstein J, Schreiber S, Bloemers MC, Bendel P, Neeman M, Schechtman E, et al. Antivascular treatment of solid melanoma tumors with bacteriochlorophyll-serine-based photodynamic therapy. Photochem Photobiol 2001 Mar;73(3):257-266.
  143. 143. Rice L, Wainwright M, Phoemix DA. Phenothiazine photosensitizers. III. Activity of methylene blue derivatives against pigmented melanoma cell lines. J Chemother 2000 Feb;12(1):94-104.
  144. 144. Busetti A, Soncin M, Reddi E, Rodgers MA, Kenney ME, Jori G. Photothermal sensitization of amelanotic melanoma cells by Ni(II)-octabutoxy-naphthalocyanine. J Photochem Photobiol B 1999 Nov-Dec;53(1-3):103-109.
  145. 145. Soncin M, Busetti A, Fusi F, Jori G, Rodgers MA. Irradiation of amelanotic melanoma cells with 532 nm high peak power pulsed laser radiation in the presence of the photothermal sensitizer Cu(II)-hematoporphyrin: a new approach to cell photoinactivation. Photochem Photobiol 1999 Jun;69(6):708-712.
  146. 146. Langer S, Abels C, Botzlar A, Pahernik S, Rick K, Szeimies RM, et al. Active and higher intracellular uptake of 5-aminolevulinic acid in tumors may be inhibited by glycine. J Invest Dermatol 1999 May;112(5):723-728.
  147. 147. Decreau R, Richard MJ, Verrando P, Chanon M, Julliard M. Photodynamic activities of silicon phthalocyanines against achromic M6 melanoma cells and healthy human melanocytes and keratinocytes. J Photochem Photobiol B 1999 Jan;48(1):48-56.
  148. 148. Busetti A, Soncin M, Jori G, Rodgers MA. High efficiency of benzoporphyrin derivative in the photodynamic therapy of pigmented malignant melanoma. Br J Cancer 1999 Feb;79(5-6):821-824.
  149. 149. Klotz LO, Fritsch C, Briviba K, Tsacmacidis N, Schliess F, Sies H. Activation of JNK and p38 but not ERK MAP kinases in human skin cells by 5-aminolevulinate-photodynamic therapy. Cancer Res 1998 Oct 1;58(19):4297-4300.
  150. 150. Busetti A, Soncin M, Jori G, Kenney ME, Rodgers MA. Treatment of malignant melanoma by high-peak-power 1064 nm irradiation followed by photodynamic therapy. Photochem Photobiol 1998 Sep;68(3):377-381.
  151. 151. Haddad R, Blumenfeld A, Siegal A, Kaplan O, Cohen M, Skornick Y, et al. In vitro and in vivo effects of photodynamic therapy on murine malignant melanoma. Ann Surg Oncol 1998 Apr-May;5(3):241-247.
  152. 152. Woodburn KW, Fan Q, Kessel D, Luo Y, Young SW. Photodynamic therapy of B16F10 murine melanoma with lutetium texaphyrin. J Invest Dermatol 1998 May;110(5):746-751.
  153. 153. Abels C, Szeimies RM, Steinbach P, Richert C, Goetz AE. Targeting of the tumor microcirculation by photodynamic therapy with a synthetic porphycene. J Photochem Photobiol B 1997 Oct;40(3):305-312.
  154. 154. Mantareva V, Shopova M, Spassova G, Wohrle D, Muller S, Jori G, et al. Si(IV)-methoxyethylene-glycol-naphthalocyanine: synthesis and pharmacokinetic and photosensitizing properties in different tumour models. J Photochem Photobiol B 1997 Oct;40(3):258-262.
  155. 155. Abels C, Fritsch C, Bolsen K, Szeimies RM, Ruzicka T, Goerz G, et al. Photodynamic therapy with 5-aminolaevulinic acid-induced porphyrins of an amelanotic melanoma in vivo. J Photochem Photobiol B 1997 Aug;40(1):76-83.
  156. 156. Kim RY, Hu LK, Flotte TJ, Gragoudas ES, Young LH. Digital angiography of experimental choroidal melanomas using benzoporphyrin derivative. Am J Ophthalmol 1997 Jun;123(6):810-816.
  157. 157. Zilberstein J, Bromberg A, Frantz A, Rosenbach-Belkin V, Kritzmann A, Pfefermann R, et al. Light-dependent oxygen consumption in bacteriochlorophyll-serine-treated melanoma tumors: on-line determination using a tissue-inserted oxygen microsensor. Photochem Photobiol 1997 Jun;65(6):1012-1019.
  158. 158. Blom DJ, Schuitmaker HJ, de Waard-Siebinga I, Dubbelman TM, Jager MJ. Decreased expression of HLA class I on ocular melanoma cells following in vitro photodynamic therapy. Cancer Lett 1997 Jan 30;112(2):239-243.
  159. 159. Fritsch C, Abels C, Goetz AE, Stahl W, Bolsen K, Ruzicka T, et al. Porphyrins preferentially accumulate in a melanoma following intravenous injection of 5-aminolevulinic acid. Biol Chem 1997 Jan;378(1):51-57.
  160. 160. Michailov N, Peeva M, Angelov I, Wohrle D, Muller S, Jori G, et al. Fluence rate effects on photodynamic therapy of B16 pigmented melanoma. J Photochem Photobiol B 1997 Jan;37(1-2):154-157.
  161. 161. Kim RY, Hu LK, Foster BS, Gragoudas ES, Young LH. Photodynamic therapy of pigmented choroidal melanomas of greater than 3-mm thickness. Ophthalmology 1996 Dec;103(12):2029-2036.
  162. 162. Hadjur C, Richard MJ, Parat MO, Jardon P, Favier A. Photodynamic effects of hypericin on lipid peroxidation and antioxidant status in melanoma cells. Photochem Photobiol 1996 Aug;64(2):375-381.
  163. 163. VanderWerf QM, Saxton RE, Chang A, Horton D, Paiva MB, Anderson J, et al. Hypericin: a new laser phototargeting agent for human cancer cells. Laryngoscope 1996 Apr;106(4):479-483.
  164. 164. Donaldson MJ, Lim L, Harper CA, Mackenzie J, G Campbell W. Primary treatment of choroidal amelanotic melanoma with photodynamic therapy. Clin Experiment Ophthalmol 2005 Oct;33(5):548-549.
  165. 165. Sheleg SV, Zhavrid EA, Khodina TV, Kochubeev GA, Istomin YP, Chalov VN, et al. Photodynamic therapy with chlorin e(6) for skin metastases of melanoma. Photodermatol Photoimmunol Photomed 2004 Feb;20(1):21-26.
  166. 166. Baldea I, Filip AG. Photodynamic therapy in melanoma--an update. J Physiol Pharmacol 2012 Apr;63(2):109-118.
  167. 167. Tuncer S, Kir N, Shields CL. Dramatic regression of amelanotic choroidal melanoma with PDT following poor response to brachytherapy. Ophthalmic Surg Lasers Imaging 2012 May 3;43(3):e38-40.
  168. 168. Canal-Fontcuberta I, Salomao DR, Robertson D, Cantrill HL, Koozekanani D, Rath PP, et al. Clinical and histopathologic findings after photodynamic therapy of choroidal melanoma. Retina 2012 May;32(5):942-948.
  169. 169. Campbell WG, Pejnovic TM. Treatment of amelanotic choroidal melanoma with photodynamic therapy. Retina 2012 Jul;32(7):1356-1362.
  170. 170. Li X, Naylor MF, Le H, Nordquist RE, Teague TK, Howard CA, et al. Clinical effects of in situ photoimmunotherapy on late-stage melanoma patients: a preliminary study. Cancer Biol Ther 2010 Dec 1;10(11):1081-1087.
  171. 171. Fasanella KE, McGrath K. Successful palliative treatment of duodenal metastatic melanoma with photodynamic therapy. Gastrointest Endosc 2009 Jan;69(1):182-184.
  172. 172. Zilidis G, Aziz F, Telara S, Eljamel MS. Fluorescence image-guided surgery and repetitive Photodynamic Therapy in brain metastatic malignant melanoma. Photodiagnosis Photodyn Ther 2008 Dec;5(4):264-266.
  173. 173. Chetty NC, Osborne V, Harland C. Amelanotic melanoma in situ: lack of sustained response to photodynamic therapy. Clin Exp Dermatol 2008 Mar;33(2):204-206.
  174. 174. Soucek P, Cihelkova I. Primary treatment of choroidal amelanotic melanoma with photodynamic therapy--comment. Clin Experiment Ophthalmol 2006 Sep-Oct;34(7):721; author reply 721-2.
  175. 175. Naylor MF, Chen WR, Teague TK, Perry LA, Nordquist RE. In situ photoimmunotherapy: a tumour-directed treatment for melanoma. Br J Dermatol 2006 Dec;155(6):1287-1292.
  176. 176. Wachtlin J, Bechrakis NE, Foerster MH. Photodynamic therapy with verteporfin for uveal melanoma. Ophthalmologe 2005 Mar;102(3):241-246.
  177. 177. Liggett PE, Lavaque AJ, Chaudhry NA, Jablon EP, Quiroz-Mercado H. Preliminary results of combined simultaneous transpupillary thermotherapy and ICG-based photodynamic therapy for choroidal melanoma. Ophthalmic Surg Lasers Imaging 2005 Nov-Dec;36(6):463-470.
  178. 178. Kubicka-Trzaska A, Starzycka M, Romanowska-Dixon B, Morawski K. Use of indocyanine green for photodynamic therapy of choroidal melanoma--preliminary report. Klin Oczna 2003;105(3-4):132-135.
  179. 179. Barbazetto IA, Lee TC, Rollins IS, Chang S, Abramson DH. Treatment of choroidal melanoma using photodynamic therapy. Am J Ophthalmol 2003 Jun;135(6):898-899.
  180. 180. Schlotzer-Schrehardt U, Viestenz A, Naumann GO, Laqua H, Michels S, Schmidt-Erfurth U. Dose-related structural effects of photodynamic therapy on choroidal and retinal structures of human eyes. Graefes Arch Clin Exp Ophthalmol 2002 Sep;240(9):748-757.
  181. 181. Favilla I, Favilla ML, Gosbell AD, Barry WR, Ellims P, Hill JS, et al. Photodynamic therapy: a 5-year study of its effectiveness in the treatment of posterior uveal melanoma, and evaluation of haematoporphyrin uptake and photocytotoxicity of melanoma cells in tissue culture. Melanoma Res 1995 Oct;5(5):355-364.
  182. 182. Chen B, Xu Y, Agostinis P, De Witte PA. Synergistic effect of photodynamic therapy with hypericin in combination with hyperthermia on loss of clonogenicity of RIF-1 cells. Int J Oncol 2001 Jun;18(6):1279-1285.
  183. 183. Wiegell SR, Fabricius S, Stender IM, Berne B, Kroon S, Andersen BL, et al. A randomized, multicentre study of directed daylight exposure times of 1(1/2) vs. 2(1/2) h in daylight-mediated photodynamic therapy with methyl aminolaevulinate in patients with multiple thin actinic keratoses of the face and scalp. Br J Dermatol 2011 May;164(5):1083-1090.
  184. 184. Alecu M, Ursaciuc C, Halalau F, Coman G, Merlevede W, Waelkens E, et al. Photodynamic treatment of basal cell carcinoma and squamous cell carcinoma with hypericin. Anticancer Res 1998 Nov-Dec;18(6B):4651-4654.
  185. 185. Bublik M, Head C, Benharash P, Paiva M, Eshraghi A, Kim T, et al. Hypericin and pulsed laser therapy of squamous cell cancer in vitro. Photomed Laser Surg 2006 Jun;24(3):341-347.
  186. 186. Chung PS, Rhee CK, Kim KH, Paek W, Chung J, Paiva MB, et al. Intratumoral hypericin and KTP laser therapy for transplanted squamous cell carcinoma. Laryngoscope 2000 Aug;110(8):1312-1316.
  187. 187. Chung PS, Saxton RE, Paiva MB, Rhee CK, Soudant J, Mathey A, et al. Hypericin uptake in rabbits and nude mice transplanted with human squamous cell carcinomas: study of a new sensitizer for laser phototherapy. Laryngoscope 1994 Dec;104(12):1471-1476.
  188. 188. Liu CD, Kwan D, Saxton RE, McFadden DW. Hypericin and photodynamic therapy decreases human pancreatic cancer in vitro and in vivo. J Surg Res 2000 Sep;93(1):137-143.
  189. 189. D'Hallewin MA, Kamuhabwa AR, Roskams T, De Witte PA, Baert L. Hypericin-based fluorescence diagnosis of bladder carcinoma. BJU Int 2002 May;89(7):760-763.
  190. 190. D'Hallewin MA, De Witte PA, Waelkens E, Merlevede W, Baert L. Fluorescence detection of flat bladder carcinoma in situ after intravesical instillation of hypericin. J Urol 2000 Aug;164(2):349-351.
  191. 191. Kubin A, Loew HG, Burner U, Jessner G, Kolbabek H, Wierrani F. How to make hypericin water-soluble. Pharmazie 2008 Apr;63(4):263-269.
  192. 192. Sim HG, Lau WK, Olivo M, Tan PH, Cheng CW. Is photodynamic diagnosis using hypericin better than white-light cystoscopy for detecting superficial bladder carcinoma? BJU Int 2005 Jun;95(9):1215-1218.
  193. 193. Kubin A, Meissner P, Wierrani F, Burner U, Bodenteich A, Pytel A, et al. Fluorescence diagnosis of bladder cancer with new water soluble hypericin bound to polyvinylpyrrolidone: PVP-hypericin. Photochem Photobiol 2008 Nov-Dec;84(6):1560-1563.
  194. 194. Bhuvaneswari R, Gan YY, Yee KK, Soo KC, Olivo M. Effect of hypericin-mediated photodynamic therapy on the expression of vascular endothelial growth factor in human nasopharyngeal carcinoma. Int J Mol Med 2007 Oct;20(4):421-428.
  195. 195. Olivo M, Du HY, Bay BH. Hypericin lights up the way for the potential treatment of nasopharyngeal cancer by photodynamic therapy. Curr Clin Pharmacol 2006 Sep;1(3):217-222.
  196. 196. Kacerovska D, Pizinger K, Majer F, Smid F. Photodynamic therapy of nonmelanoma skin cancer with topical hypericum perforatum extract--a pilot study. Photochem Photobiol 2008 May-Jun;84(3):779-785.
  197. 197. Davids LM, Kleemann B, Cooper S, Kidson SH. Melanomas display increased cytoprotection to hypericin-mediated cytotoxicity through the induction of autophagy. Cell Biol Int 2009 Oct;33(10):1065-1072.
  198. 198. Mosmann T. Rapid colorimetric assay for cellular growth and survival: application to proliferation and cytotoxicity assays. J Immunol Methods 1983 Dec 16;65(1-2):55-63.
  199. 199. Cory AH, Owen TC, Barltrop JA, Cory JG. Use of an aqueous soluble tetrazolium/formazan assay for cell growth assays in culture. Cancer Commun 1991 Jul;3(7):207-212.
  200. 200. Davids LM, Kleemann B. Combating melanoma: The use of photodynamic therapy as a novel, adjuvant therapeutic tool. Cancer Treat Rev 2010 Dec 16.
  201. 201. Davids LM, du Toit E, Kidson SH, Todd G. A rare repigmentation pattern in a vitiligo patient: a clue to an epidermal stem-cell reservoir of melanocytes? Clin Exp Dermatol 2009 Mar;34(2):246-248.
  202. 202. Kroemer G, El-Deiry WS, Golstein P, Peter ME, Vaux D, Vandenabeele P, et al. Classification of cell death: recommendations of the Nomenclature Committee on Cell Death. Cell Death Differ 2005 Nov;12 Suppl 2:1463-1467.
  203. 203. Golstein P, Kroemer G. Cell death by necrosis: towards a molecular definition. Trends Biochem Sci 2007 Jan;32(1):37-43.
  204. 204. Galluzzi L, Vitale I, Abrams JM, Alnemri ES, Baehrecke EH, Blagosklonny MV, et al. Molecular definitions of cell death subroutines: recommendations of the Nomenclature Committee on Cell Death 2012. Cell Death Differ 2012 Jan;19(1):107-120.
  205. 205. Hanahan D, Weinberg RA. Hallmarks of cancer: the next generation. Cell 2011 Mar 4;144(5):646-674.
  206. 206. Wellbrock C, Weisser C, Geissinger E, Troppmair J, Schartl M. Activation of p59(Fyn) leads to melanocyte dedifferentiation by influencing MKP-1-regulated mitogen-activated protein kinase signaling. J Biol Chem 2002 Feb 22;277(8):6443-6454.
  207. 207. Cohen C, Zavala-Pompa A, Sequeira JH, Shoji M, Sexton DG, Cotsonis G, et al. Mitogen-actived protein kinase activation is an early event in melanoma progression. Clin Cancer Res 2002 Dec;8(12):3728-3733.
  208. 208. Tsao H, Goel V, Wu H, Yang G, Haluska FG. Genetic interaction between NRAS and BRAF mutations and PTEN/MMAC1 inactivation in melanoma. J Invest Dermatol 2004 Feb;122(2):337-341.
  209. 209. Davies H, Bignell GR, Cox C, Stephens P, Edkins S, Clegg S, et al. Mutations of the BRAF gene in human cancer. Nature 2002 Jun 27;417(6892):949-954.
  210. 210. Gray-Schopfer VC, da Rocha Dias S, Marais R. The role of B-RAF in melanoma. Cancer Metastasis Rev 2005 Jan;24(1):165-183.
  211. 211. Tong Z, Singh G, Rainbow AJ. Sustained activation of the extracellular signal-regulated kinase pathway protects cells from photofrin-mediated photodynamic therapy. Cancer Res 2002 Oct 1;62(19):5528-5535.
  212. 212. Mercurio F, Manning AM. NF-kappaB as a primary regulator of the stress response. Oncogene 1999 Nov 1;18(45):6163-6171.
  213. 213. Ryter SW, Gomer CJ. Nuclear factor kappa B binding activity in mouse L1210 cells following photofrin II-mediated photosensitization. Photochem Photobiol 1993 Nov;58(5):753-756.
  214. 214. Danial NN, Korsmeyer SJ. Cell death: critical control points. Cell 2004 Jan 23;116(2):205-219.
  215. 215. Oleinick NL, Morris RL, Belichenko I. The role of apoptosis in response to photodynamic therapy: what, where, why, and how. Photochem Photobiol Sci 2002 Jan;1(1):1-21.
  216. 216. Agostinis P, Buytaert E, Breyssens H, Hendrickx N. Regulatory pathways in photodynamic therapy induced apoptosis. Photochem Photobiol Sci 2004 Aug;3(8):721-729.
  217. 217. Buytaert E, Callewaert G, Vandenheede JR, Agostinis P. Deficiency in apoptotic effectors Bax and Bak reveals an autophagic cell death pathway initiated by photodamage to the endoplasmic reticulum. Autophagy 2006 Jul-Sep;2(3):238-240.
  218. 218. Fisher DE. Apoptosis in cancer therapy: crossing the threshold. Cell 1994 Aug 26;78(4):539-542.
  219. 219. Micheau O, Solary E, Hammann A, Martin F, Dimanche-Boitrel MT. Sensitization of cancer cells treated with cytotoxic drugs to fas-mediated cytotoxicity. J Natl Cancer Inst 1997 Jun 4;89(11):783-789.
  220. 220. Ali SM, Chee SK, Yuen GY, Olivo M. Hypericin induced death receptor-mediated apoptosis in photoactivated tumor cells. Int J Mol Med 2002 Jun;9(6):601-616.
  221. 221. Reiners JJ,Jr, Agostinis P, Berg K, Oleinick NL, Kessel D. Assessing autophagy in the context of photodynamic therapy. Autophagy 2010 Jan;6(1):7-18.
  222. 222. Dewaele M, Maes H, Agostinis P. ROS-mediated mechanisms of autophagy stimulation and their relevance in cancer therapy. Autophagy 2010 Oct 19;6(7).
  223. 223. Garg AD, Krysko DV, Vandenabeele P, Agostinis P. DAMPs and PDT-mediated photo-oxidative stress: exploring the unknown. Photochem Photobiol Sci 2011 May;10(5):670-680.
  224. 224. Garg AD, Nowis D, Golab J, Vandenabeele P, Krysko DV, Agostinis P. Immunogenic cell death, DAMPs and anticancer therapeutics: an emerging amalgamation. Biochim Biophys Acta 2010 Jan;1805(1):53-71.
  225. 225. Green DR, Ferguson T, Zitvogel L, Kroemer G. Immunogenic and tolerogenic cell death. Nat Rev Immunol 2009 May;9(5):353-363.
  226. 226. Zitvogel L, Kepp O, Kroemer G. Decoding cell death signals in inflammation and immunity. Cell 2010 Mar 19;140(6):798-804.
  227. 227. Zitvogel L, Kepp O, Senovilla L, Menger L, Chaput N, Kroemer G. Immunogenic tumor cell death for optimal anticancer therapy: the calreticulin exposure pathway. Clin Cancer Res 2010 Jun 15;16(12):3100-3104.
  228. 228. Demaria S, Pikarsky E, Karin M, Coussens LM, Chen YC, El-Omar EM, et al. Cancer and inflammation: promise for biologic therapy. J Immunother 2010 May;33(4):335-351.
  229. 229. Garg AD, Nowis D, Golab J, Agostinis P. Photodynamic therapy: illuminating the road from cell death towards anti-tumour immunity. Apoptosis 2010 Mar 10.
  230. 230. Buytaert E, Callewaert G, Hendrickx N, Scorrano L, Hartmann D, Missiaen L, et al. Role of endoplasmic reticulum depletion and multidomain proapoptotic BAX and BAK proteins in shaping cell death after hypericin-mediated photodynamic therapy. FASEB J 2006 Apr;20(6):756-758.
  231. 231. Ritz R, Roser F, Radomski N, Strauss WS, Tatagiba M, Gharabaghi A. Subcellular colocalization of hypericin with respect to endoplasmic reticulum and Golgi apparatus in glioblastoma cells. Anticancer Res 2008 Jul-Aug;28(4B):2033-2038.
  232. 232. Galanou MC, Theodossiou TA, Tsiourvas D, Sideratou Z, Paleos CM. Interactive transport, subcellular relocation and enhanced phototoxicity of hypericin encapsulated in guanidinylated liposomes via molecular recognition. Photochem Photobiol 2008 Sep-Oct;84(5):1073-1083.
  233. 233. Ali SM, Olivo M. Bio-distribution and subcellular localization of Hypericin and its role in PDT induced apoptosis in cancer cells. Int J Oncol 2002 Sep;21(3):531-540.
  234. 234. Ali SM, Chee SK, Yuen GY, Olivo M. Hypericin induced death receptor-mediated apoptosis in photoactivated tumor cells. Int J Mol Med 2002 Jun;9(6):601-616.
  235. 235. English DS, Doyle RT, Petrich JW, Haydon PG. Subcellular distributions and excited-state processes of hypericin in neurons. Photochem Photobiol 1999 Mar;69(3):301-305.
  236. 236. Crnolatac I, Huygens A, Agostinis P, Kamuhabwa AR, Maes J, van Aerschot A, et al. In vitro accumulation and permeation of hypericin and lipophilic analogues in 2-D and 3-D cellular systems. Int J Oncol 2007 Feb;30(2):319-324.
  237. 237. Theodossiou TA, Hothersall JS, De Witte PA, Pantos A, Agostinis P. The Multifaceted Photocytotoxic Profile of Hypericin. Mol Pharm 2009 Sep 24.
  238. 238. Ho YF, Wu MH, Cheng BH, Chen YW, Shih MC. Lipid-mediated preferential localization of hypericin in lipid membranes. Biochim Biophys Acta 2009 Jun;1788(6):1287-1295.
  239. 239. Mojzisova H, Bonneau S, Brault D. Structural and physico-chemical determinants of the interactions of macrocyclic photosensitizers with cells. Eur Biophys J 2007 Nov;36(8):943-953.
  240. 240. Buytaert E, Matroule JY, Durinck S, Close P, Kocanova S, Vandenheede JR, et al. Molecular effectors and modulators of hypericin-mediated cell death in bladder cancer cells. Oncogene 2008 Mar 20;27(13):1916-1929.
  241. 241. Verfaillie T, Salazar M, Velasco G, Agostinis P. Linking ER Stress to Autophagy: Potential Implications for Cancer Therapy. Int J Cell Biol 2010;2010:930509.
  242. 242. Thomas C, MacGill RS, Miller GC, Pardini RS. Photoactivation of hypericin generates singlet oxygen in mitochondria and inhibits succinoxidase. Photochem Photobiol 1992 Jan;55(1):47-53.
  243. 243. Miccoli L, Beurdeley-Thomas A, De Pinieux G, Sureau F, Oudard S, Dutrillaux B, et al. Light-induced photoactivation of hypericin affects the energy metabolism of human glioma cells by inhibiting hexokinase bound to mitochondria. Cancer Res 1998 Dec 15;58(24):5777-5786.
  244. 244. Theodossiou TA, Papakyriakou A, Hothersall JS. Molecular modeling and experimental evidence for hypericin as a substrate for mitochondrial complex III; mitochondrial photodamage as demonstrated using specific inhibitors. Free Radic Biol Med 2008 Dec 1;45(11):1581-1590.
  245. 245. Utsumi T, Okuma M, Kanno T, Takehara Y, Yoshioka T, Fujita Y, et al. Effect of the antiretroviral agent hypericin on rat liver mitochondria. Biochem Pharmacol 1995 Aug 25;50(5):655-662.
  246. 246. Buytaert E, Callewaert G, Vandenheede JR, Agostinis P. Deficiency in apoptotic effectors Bax and Bak reveals an autophagic cell death pathway initiated by photodamage to the endoplasmic reticulum. Autophagy 2006 Jul-Sep;2(3):238-240.
  247. 247. Castano AP, Mroz P, Hamblin MR. Photodynamic therapy and anti-tumour immunity. Nat Rev Cancer 2006 Jul;6(7):535-545.
  248. 248. Panaretakis T, Kepp O, Brockmeier U, Tesniere A, Bjorklund AC, Chapman DC, et al. Mechanisms of pre-apoptotic calreticulin exposure in immunogenic cell death. EMBO J 2009 Mar 4;28(5):578-590.
  249. 249. Young AR, Narita M, Ferreira M, Kirschner K, Sadaie M, Darot JF, et al. Autophagy mediates the mitotic senescence transition. Genes Dev 2009 Apr 1;23(7):798-803.
  250. 250. Lu Z, Luo RZ, Lu Y, Zhang X, Yu Q, Khare S, et al. The tumor suppressor gene ARHI regulates autophagy and tumor dormancy in human ovarian cancer cells. J Clin Invest 2008 Dec;118(12):3917-3929.
  251. 251. Gewirtz DA. Autophagy, senescence and tumor dormancy in cancer therapy. Autophagy 2009 Nov;5(8):1232-1234.
  252. 252. Nystrom T. Role of oxidative carbonylation in protein quality control and senescence. EMBO J 2005 Apr 6;24(7):1311-1317.
  253. 253. Pattingre S, Tassa A, Qu X, Garuti R, Liang XH, Mizushima N, et al. Bcl-2 antiapoptotic proteins inhibit Beclin 1-dependent autophagy. Cell 2005 Sep 23;122(6):927-939.
  254. 254. Coussens LM, Werb Z. Inflammation and cancer. Nature 2002 Dec 19-26;420(6917):860-867.
  255. 255. Szatrowski TP, Nathan CF. Production of large amounts of hydrogen peroxide by human tumor cells. Cancer Res 1991 Feb 1;51(3):794-798.
  256. 256. Toyokuni S, Okamoto K, Yodoi J, Hiai H. Persistent oxidative stress in cancer. FEBS Lett 1995 Jan 16;358(1):1-3.
  257. 257. Sander CS, Chang H, Hamm F, Elsner P, Thiele JJ. Role of oxidative stress and the antioxidant network in cutaneous carcinogenesis. Int J Dermatol 2004 May;43(5):326-335.
  258. 258. Lincoln DT, Ali Emadi EM, Tonissen KF, Clarke FM. The thioredoxin-thioredoxin reductase system: over-expression in human cancer. Anticancer Res 2003 May-Jun;23(3B):2425-2433.
  259. 259. Cen D, Gonzalez RI, Buckmeier JA, Kahlon RS, Tohidian NB, Meyskens FL,Jr. Disulfiram induces apoptosis in human melanoma cells: a redox-related process. Mol Cancer Ther 2002 Jan;1(3):197-204.
  260. 260. Cen D, Brayton D, Shahandeh B, Meyskens FL,Jr, Farmer PJ. Disulfiram facilitates intracellular Cu uptake and induces apoptosis in human melanoma cells. J Med Chem 2004 Dec 30;47(27):6914-6920.
  261. 261. Huang P, Feng L, Oldham EA, Keating MJ, Plunkett W. Superoxide dismutase as a target for the selective killing of cancer cells. Nature 2000 Sep 21;407(6802):390-395.
  262. 262. Dobos J, Timar J, Bocsi J, Burian Z, Nagy K, Barna G, et al. In vitro and in vivo antitumor effect of 2-methoxyestradiol on human melanoma. Int J Cancer 2004 Dec 10;112(5):771-776.
  263. 263. Russo GL. Ins and outs of dietary phytochemicals in cancer chemoprevention. Biochem Pharmacol 2007 Aug 15;74(4):533-544.
  264. 264. Tyagi AK, Singh RP, Agarwal C, Chan DC, Agarwal R. Silibinin strongly synergizes human prostate carcinoma DU145 cells to doxorubicin-induced growth Inhibition, G2-M arrest, and apoptosis. Clin Cancer Res 2002 Nov;8(11):3512-3519.
  265. 265. De Larco JE, Park CA, Dronava H, Furcht LT. Paradoxical roles for antioxidants in tumor prevention and eradication. Cancer Biol Ther 2010 Mar 8;9(5).
  266. 266. Farmer PJ, Gidanian S, Shahandeh B, Di Bilio AJ, Tohidian N, Meyskens FL,Jr. Melanin as a target for melanoma chemotherapy: pro-oxidant effect of oxygen and metals on melanoma viability. Pigment Cell Res 2003 Jun;16(3):273-279.
  267. 267. Chen KG, Valencia JC, Lai B, Zhang G, Paterson JK, Rouzaud F, et al. Melanosomal sequestration of cytotoxic drugs contributes to the intractability of malignant melanomas. Proc Natl Acad Sci U S A 2006 Jun 27;103(26):9903-9907.
  268. 268. Chen KG, Leapman RD, Zhang G, Lai B, Valencia JC, Cardarelli CO, et al. Influence of melanosome dynamics on melanoma drug sensitivity. J Natl Cancer Inst 2009 Sep 16;101(18):1259-1271.
  269. 269. Raposo G, Marks MS. The dark side of lysosome-related organelles: specialization of the endocytic pathway for melanosome biogenesis. Traffic 2002 Apr;3(4):237-248.
  270. 270. Hearing VJ. Biogenesis of pigment granules: a sensitive way to regulate melanocyte function. J Dermatol Sci 2005 Jan;37(1):3-14.
  271. 271. Baldea I. Melanocyte pigmentation – friend or foe on the route to melanoma. In: Tanaka Y, editor. Breakthroughs in Melanoma Research Rijeka, Croatia: InTech; 2011. p. 183-217.
  272. 272. Solano F, Martinez-Esparza M, Jimenez-Cervantes C, Hill SP, Lozano JA, Garcia-Borron JC. New insights on the structure of the mouse silver locus and on the function of the silver protein. Pigment Cell Res 2000;13 Suppl 8:118-124.
  273. 273. Kwon BS, Chintamaneni C, Kozak CA, Copeland NG, Gilbert DJ, Jenkins N, et al. A melanocyte-specific gene, Pmel 17, maps near the silver coat color locus on mouse chromosome 10 and is in a syntenic region on human chromosome 12. Proc Natl Acad Sci U S A 1991 Oct 15;88(20):9228-9232.
  274. 274. Theos AC, Truschel ST, Raposo G, Marks MS. The Silver locus product Pmel17/gp100/Silv/ME20: controversial in name and in function. Pigment Cell Res 2005 Oct;18(5):322-336.
  275. 275. Orlow SJ. Melanosomes are specialized members of the lysosomal lineage of organelles. J Invest Dermatol 1995 Jul;105(1):3-7.
  276. 276. Raposo G, Tenza D, Murphy DM, Berson JF, Marks MS. Distinct protein sorting and localization to premelanosomes, melanosomes, and lysosomes in pigmented melanocytic cells. J Cell Biol 2001 Feb 19;152(4):809-824.
  277. 277. Valencia JC, Hoashi T, Pawelek JM, Solano F, Hearing VJ. Pmel17: controversial indeed but critical to melanocyte function. Pigment Cell Res 2006 Jun;19(3):250-2; author reply 253-7.
  278. 278. Nelson JS, McCullough JL, Berns MW. Photodynamic therapy of human malignant melanoma xenografts in athymic nude mice. J Natl Cancer Inst 1988 Mar 2;80(1):56-60.
  279. 279. Meyskens FL,Jr, Farmer P, Fruehauf JP. Redox regulation in human melanocytes and melanoma. Pigment Cell Res 2001 Jun;14(3):148-154.
  280. 280. Shackleton M, Quintana E, Fearon ER, Morrison SJ. Heterogeneity in cancer: cancer stem cells versus clonal evolution. Cell 2009 Sep 4;138(5):822-829.
  281. 281. Marotta LL, Polyak K. Cancer stem cells: a model in the making. Curr Opin Genet Dev 2009 Feb;19(1):44-50.
  282. 282. Al-Hajj M, Wicha MS, Benito-Hernandez A, Morrison SJ, Clarke MF. Prospective identification of tumorigenic breast cancer cells. Proc Natl Acad Sci U S A 2003 Apr 1;100(7):3983-3988.
  283. 283. Bonnet D, Dick JE. Human acute myeloid leukemia is organized as a hierarchy that originates from a primitive hematopoietic cell. Nat Med 1997 Jul;3(7):730-737.
  284. 284. O'Brien CA, Pollett A, Gallinger S, Dick JE. A human colon cancer cell capable of initiating tumour growth in immunodeficient mice. Nature 2007 Jan 4;445(7123):106-110.
  285. 285. Singh SK, Hawkins C, Clarke ID, Squire JA, Bayani J, Hide T, et al. Identification of human brain tumour initiating cells. Nature 2004 Nov 18;432(7015):396-401.
  286. 286. Abel EV, Aplin AE. Finding the root of the problem: the quest to identify melanoma stem cells. Front Biosci (Schol Ed) 2011 Jun 1;3:937-945.
  287. 287. Jones PM, George AM. The ABC transporter structure and mechanism: perspectives on recent research. Cell Mol Life Sci 2004 Mar;61(6):682-699.
  288. 288. Frank NY, Margaryan A, Huang Y, Schatton T, Waaga-Gasser AM, Gasser M, et al. ABCB5-mediated doxorubicin transport and chemoresistance in human malignant melanoma. Cancer Res 2005 May 15;65(10):4320-4333.
  289. 289. Keshet GI, Goldstein I, Itzhaki O, Cesarkas K, Shenhav L, Yakirevitch A, et al. MDR1 expression identifies human melanoma stem cells. Biochem Biophys Res Commun 2008 Apr 18;368(4):930-936.
  290. 290. Monzani E, Facchetti F, Galmozzi E, Corsini E, Benetti A, Cavazzin C, et al. Melanoma contains CD133 and ABCG2 positive cells with enhanced tumourigenic potential. Eur J Cancer 2007 Mar;43(5):935-946.
  291. 291. Robey RW, Steadman K, Polgar O, Bates SE. ABCG2-mediated transport of photosensitizers: potential impact on photodynamic therapy. Cancer Biol Ther 2005 Feb;4(2):187-194.
  292. 292. Tamura A, An R, Hagiya Y, Hoshijima K, Yoshida T, Mikuriya K, et al. Drug-induced phototoxicity evoked by inhibition of human ABC transporter ABCG2: development of in vitro high-speed screening systems. Expert Opin Drug Metab Toxicol 2008 Mar;4(3):255-272.
  293. 293. Liu W, Baer MR, Bowman MJ, Pera P, Zheng X, Morgan J, et al. The tyrosine kinase inhibitor imatinib mesylate enhances the efficacy of photodynamic therapy by inhibiting ABCG2. Clin Cancer Res 2007 Apr 15;13(8):2463-2470.

Written By

L.M. Davids and B. Kleemann

Submitted: 31 August 2012 Published: 30 January 2013