The in vivo efficacy of phthalocyanine–nanoparticle conjugates for the photodynamic therapy of amelanotic melanoma

https://doi.org/10.1016/j.ejca.2010.02.037Get rights and content

Abstract

The efficiency of a Zn(II)-phthalocyanine disulphide (C11Pc), a compound with both phthalocyanine units bearing seven hexyl chains and a sulphur terminated C11 chain, as a photodynamic therapy (PDT) agent was investigated in C57 mice bearing a sub-cutaneously transplanted amelanotic melanoma. The phthalocyanine was intravenously injected at a dose of 1.5 μmol/kg body weight either free or bound to gold nanoparticles, using a Cremophor emulsion as a delivery vehicle. Biodistribution studies at selected post-injection times showed that the nanoparticle-associated C11Pc was recovered in significantly larger amounts from all the examined tissues and the serum and yielded a greater selectivity of tumour targeting: thus, the ratio between the amount of phthalocyanine recovered from the amelanotic melanoma and the skin (peritumoural tissue) increased from 2.3 to 5.5 from the free to the gold nanoparticle-bound C11Pc at 24 h after injection. PDT studies with the C11Pc-loaded amelanotic melanoma showed a markedly more significant response of the tumour in the mice that had received the nanoparticle-bound photosensitiser; the PDT effect was especially extensive if the irradiation was performed at 3 h after C11Pc injection when large phthalocyanine amounts were still present in the serum. This suggests that the PDT promoted by C11Pc predominantly acts via vascular damage at least in this specific animal model. This hypothesis was fully confirmed by electron microscopy observations of tumour specimens obtained at different times after the end of PDT, showing an extensive damage of the blood capillaries and the endothelial cells.

Introduction

Photodynamic therapy (PDT) is a well-established modality for the treatment of localised tumours which is used in clinical practice as an alternative or adjuvant to conventional therapies, such as radiotherapy, surgery or chemotherapy. PDT selectively destroys neoplastic lesions by the combined action of a light-activated drug, termed a photosensitiser, and visible light. The wavelengths of the light that are typically used for PDT are in the red or near infrared spectral range as these wavelengths exhibit a greater penetration depth into most human tissues and are not absorbed by normal tissue constituents. Upon irradiation, the photosensitiser is promoted to the long-lived lowest excited triplet state. The energy of the excited state photosensitiser molecule is transferred to the ground state of oxygen to produce the singlet state oxygen species. Singlet oxygen is cytotoxic and it is the production of this species which results in the destruction of cancerous tissue.1 Alternatively, the triplet photosensitiser can promote electron transfer processes with nearby substrates with the generation of radical species (including the superoxide anion and the hydroxyl radical), which can in turn induce different types of damage in the microenvironment of the photosensitiser binding site.1

Approval of PDT for oncological indications was first granted in 1993. Currently four drugs have received regulatory approval in North America and/or the European Union2: Photofrin (a complex mixture of haematoporphyrin derivatives, Axcan Pharma Inc., Mont-Saint-Hilaire, Canada), for advanced and early lung cancer, superficial gastric cancer, oesophageal adenocarcinoma, cervical cancer and bladder cancer; Foscan (meta-tetrahydroxyphenyl chlorin, Biolitec Pharma Ltd., Dublin, Ireland), for palliative head and neck cancer; the other two drugs, Levulan (5-aminolevulinic acid; ALA, Dusa Pharmaceuticals Inc., Wilmington, MA, USA) and Metvix (methyl 5-aminolevulinate, Photocure ASA, Oslo, Norway), are not themselves photoactive but when applied to basal cell carcinoma, for example, are metabolically converted to protoporphyrin IX (or a methyl ester derivative for Metvix) via the haem biosynthetic pathway. Protoporphyrin IX is a photosensitiser and consequently produces singlet oxygen when stimulated by light. Photodynamic therapy with these four photosensitiser drugs has been used to treat several thousand patients worldwide.3, 4 While excellent clinical results are achieved, the full exploitation of the potential of PDT is impeded by factors such as the somewhat limited selectivity of tumour targeting; the prolonged persistence of the available PDT agents in the skin, causing a generalised cutaneous photosensitivity; and the chemical heterogeneity and inefficient red light absorption typical of Photofrin and ALA-derived protoporphyrin.1, 3 The clinical status of photodynamic therapy for cancer treatment has been recently reviewed.2, 3, 4

Novel approaches are being investigated in order to overcome the present limitations of PDT, including the development of second-generation photosensitisers with improved photochemical and tumour-localising properties. Many of the compounds developed as second-generation photosensitisers have limited water solubility as hydrophobic photosensitisers typically exhibit a better tumour targeting and substantially greater PDT efficacy.1, 5 The obvious limitation of such water-insoluble photosensitisers is that they require a vehicle to deliver the drug to the tumour tissue.

While numerous drug delivery vehicles exist, particularly encouraging results have been obtained recently which suggest that nanoparticle-carried PDT agents are accumulated in significant amounts by a variety of tumour cells.6, 7, 8, 9 Upon accumulation in the tumour and subsequent activation by suitable visible-light wavelengths, these photosensitiser–nanoparticle conjugates induce an efficient damage of the malignant tissue.9, 10, 11 Photophysical investigations have demonstrated that the association of a photosensitiser with nanoparticles does not necessarily alter its excited state properties nor the efficiency of singlet oxygen generation.10, 11, 12 Thus, PDT could take advantage of the increasing evidence supporting the concept that nanoparticles have a large potential to act as a viable enhancer of tumour targeting by anti-neoplastic drugs.13, 14

Many different materials have been used to formulate nanoparticles for drug delivery. Various photosensitisers, many of which are hydrophobic, have been encapsulated within water-soluble polymers such as polylactic-co-glycolic acid and polylactic acid, to overcome the problems associated with solubilisation. For example, meso-tetra(4-hydroxyphenyl)porphyrin,15, 16 bacteriochlorophyll-a,17 verteporfin,18 methylene blue,19 hypericin20 and various phthalocyanines21, 22, 23 have all been encapsulated in such water-soluble polymers. Silica nanoparticles have been used successfully for the delivery of the readily available PDT photosensitisers, such as pheophorbides and chlorins,7, 24 or with unusual sensitisers, such as fullerene.25 Cyclodextrin-based nanoparticles also have been used to entrap photosensitisers and shown to exhibit good PDT efficiency.26, 27

We28, 29 and others30 have developed gold nanoparticles as carriers of hydrophobic phthalocyanines, examples of second-generation photosensitisers. In this instance, rather than the photosensitiser being encapsulated within the nanoparticle carrier, the photosensitiser is bound to the surface of the gold nanoparticles via a thiol tether specifically designed so that the molecule will form a self-assembled monolayer on the gold nanoparticle surface.28 The formation of the phthalocyanine on the gold nanoparticle surface has the potential advantage that the hydrophobic character of the photosensitiser is maintained so that the conjugates retain the enhanced tumour targeting and greater PDT efficacy. It has also been suggested that the binding of the photosensitiser to the surface of the nanoparticle may represent an advantage as singlet oxygen does not need to diffuse out of the polymeric/silica nanoparticle structure as per encapsulated photosensitisers.11

While the in vitro results are extremely promising, to date there are only a limited number of studies in which the in vivo efficacy of photosensitiser–nanoparticle conjugates has been investigated.18, 30, 31, 32, 33, 34, 35 In this paper, we describe the pharmacokinetic behaviour and PDT efficacy of phthalocyanine–gold nanoparticle conjugates for the treatment of a sub-cutaneously implanted amelanotic melanoma. This tumour was selected as a model for the present investigations since it is a common skin tumour that favourably responds to PDT treatment36 and it has been repeatedly adopted for assessing the efficacy of the experimental PDT with a number of photosensitising agents.37, 38

Section snippets

Photosensitising agents

The following photosensitising agents were used in the pharmacokinetic and experimental PDT studies: 1,1′,4,4′,8,8′,15,15′,18,18′,22,22′-tetradecakisdecyl-25,25′-(11,11′dithiodiundecyl)diphthalocyanine (C11Pc), (the free, non-bound, phthalocyanine photosensitiser) and the nanoconstruct made by conjugating the C11Pc photosensitiser with gold nanoparticles (C11Pc-Np). The chemical structure of the C11Pc is shown in Fig. 1. The synthesis of C11Pc and the formulation of the C11Pc-Np nanoparticle

Pharmacokinetic studies with C11Pc and the C11Pc-Np conjugate

The time-dependent distribution of the i.v.-injected C11Pc in C57 mice bearing a sub-cutaneously transplanted amelanotic melanoma (Fig. 2A) is similar to that previously determined for structurally analogous octa-decyl or octa-pentyl phthalocyanine derivatives albeit in a different animal model.40, 41 Thus, the largest accumulation of the photosensitising agent was found to occur in the constituents of the reticulo-endothelial system, such as liver and spleen. This observation is in agreement

Discussion

The utilisation of delivery vehicles for the administration of photodynamic agents by systemic routes has been the subject of active debate and several experimental studies among both basic investigators and clinical users in the field of PDT.43 In practice, the photosensitisers which usually exhibit the largest affinity for tumour tissues are characterised by a relatively high degree of hydrophobicity, hence their direct intravenous injection into the bloodstream is greatly facilitated by

Conclusions

The pharmacokinetic data reveal that by conjugating the C11Pc photosensitiser to gold nanoparticles an enhanced accumulation within a sub-cutaneously implanted amelanotic melanoma is achieved as compared with the free phthalocyanine. Photodynamic therapy studies confirm that tumour growth is optimally slowed following light treatment 3 h post-i.v. injection. Electron microscopy studies have confirmed that the mechanism of photodamage of the tumour is via destruction of the vasculature. The data

Conflict of interest statement

None declared.

Acknowledgement

Funding from Cancer Research UK (Grant C22031/A7097) to DAR and GJ which supported this work is gratefully acknowledged.

References (52)

  • N. Nishiyama et al.

    Enhanced photodynamic cancer treatment by supramolecular nanocarriers charged with dendrimer phthalocyanine

    J Control Release

    (2009)
  • M. Soncin et al.

    Pharmacokinetic and phototherapeutic properties of axially substituted Si(IV)-tetradibenzobarreleno-octabutoxyphthalocyanines

    J Photochem Photobiol B: Biol

    (1997)
  • G. Jori

    Tumour photosensitisers: approaches to enhance the selectivity and efficiency of photodynamic therapy

    J Photochem Photobiol B: Biol

    (1996)
  • J.S. Friedberg et al.

    A phase I study of Foscan-mediated photodynamic therapy in patients with mesothelioma

    Ann Thorac Surg

    (2003)
  • A.P. Castano et al.

    Mechanisms in photodynamic therapy. 2. Cellular signalling, cell metabolism and modes of cell death

    Photodiag Photodyn Ther

    (2005)
  • W.M. Sharman et al.

    Targeted photodynamic therapy via receptor mediated delivery systems

    Adv Drug Deliv Rev

    (2004)
  • J.K. Gbadamosi et al.

    Pegylation of microspheres generates a heterogeneous population of particles with different surface characteristics and biological performance

    FEBS Lett

    (2002)
  • T.J. Dougherty et al.

    Photodynamic therapy

    J Natl Cancer Inst

    (1998)
  • M. Triesscheijn et al.

    Photodynamic therapy in oncology

    The Oncologist

    (2000)
  • A. Juzeniene et al.

    Milestones in the development of photodynamic therapy and fluorescence diagnosis

    Photochem Photobiol Sci

    (2007)
  • A.E. O’Connor et al.

    Porphyrin and Nonporphyrin photosensitisers in oncology: preclinical and clinical advances in photodynamic therapy

    Photochem Photobiol

    (2009)
  • R.W. Boyle et al.

    Structure and biodistribution relationships of photodynamic sensitizers

    Photochem Photobiol

    (1996)
  • L.O. Cinteza et al.

    Diacyllipid micelle-based nanocarrier for magnetically guided delivery of drugs in photodynamic therapy

    Mol Pharm

    (2006)
  • I. Roy et al.

    Ceramic-based nanoparticles entrapping water-insoluble photosensitising anticancer drugs: novel drug-carrier system for photodynamic therapy

    J Am Chem Soc

    (2003)
  • S.Z. Wang et al.

    Nanomaterials and singlet oxygen photosensitisers: potential applications in photodynamic therapy

    J Mater Chem

    (2004)
  • D. Gao et al.

    Nanoparticles for two-photon photodynamic therapy in living cells

    Nano Lett

    (2006)
  • Cited by (150)

    View all citing articles on Scopus
    View full text