Photodynamic therapy as an antifungal treatment (Review)

  • Authors:
    • Yi Liang
    • Li‑Ming Lu
    • Yong Chen
    • You‑Kun Lin
  • View Affiliations

  • Published online on: May 11, 2016     https://doi.org/10.3892/etm.2016.3336
  • Pages: 23-27
Metrics: Total Views: 0 (Spandidos Publications: | PMC Statistics: )
Total PDF Downloads: 0 (Spandidos Publications: | PMC Statistics: )


Abstract

Photodynamic therapy (PDT) involves the systemic or topical application of a photosensitizer (PS), alongside the selective illumination of the target lesion with light of an appropriate wavelength, in order to promote localized oxidative photodamage and subsequent cell death. Numerous studies have demonstrated that PDT is highly effective in the destruction of fungi in vitro. The mechanism underlying the effects of PDT results from the photons of visible light of an appropriate wavelength interacting with the intracellular molecules of the PS. Reactive species are produced as a result of the oxidative stress caused by the interaction between the visible light and the biological tissue. At present, no antifungal treatment based on PDT has been licensed. However, antifungal PDT is emerging as an area of interest for research.

Introduction

Photodynamic therapy (PDT) involves the systemic or topical administration of a photosensitizer (PS), alongside the selective illumination of a target lesion with light of an appropriate wavelength, in order to promote localized oxidative photodamage and subsequent cell death (1,2). PDT showed initial success in the treatment of malignant diseases, including skin tumors (3), cutaneous T-cell lymphoma (4) and cervical cancer (5,6), and precancerous lesions, including Bowen's disease (7,8) and Barrett's oesophagus (9). In recent years, PDT has also been used for the treatment of vulgaris, leishmaniasis, acne and bacterial, fungal and viral infections (1012).

Previous studies demonstrated that PDT was highly effective in the destruction of fungi in vitro (13,14). At present, numerous antifungal drugs, including azoles, have a fungistatic (a delay in growth) rather than fungicidal (a complete inactivation of fungal conidia and hyphae) effect. Fungal conidia have been shown to be less susceptible to antifungal drugs, compared with hyphae (10,15).

It is estimated that 10–20% of the global population may be affected by mycoses, which are frequently recurrent and chronic (16). Acquisition of fungal pathogens results in significant morbidity causing discomfort, social isolation, disfigurement and may predispose one to bacterial diseases (17,18). However, fungi are eukaryotic organisms and their similarities to mammalian cells have led to significant difficulties in the development of new antifungal drugs. The heavy burden of fungal infections, and the increase in fungal strains resistant to the current antifungals globally (18), has rendered the development of new therapeutic strategies, such as antifungal photodynamic therapy, an urgent requirement.

PDT

PDT uses a PS and visible light of the appropriate wavelength to generate cytotoxic reactive species in the presence of oxygen. The presence of cytotoxic species in the target site results in the damage of target cells (19). PDT involves delivering visible light of the appropriate wavelength to excite the PS molecule to the excited singlet state (19).

The primary advantages of PDT are that the PS can be targeted to a specific cell or tissue and the visible light can be spatially directed to the infected area (19). In addition, the treatment of localized infections with PDT allows selectivity of the PS for microbes over host cells, delivery of the PS into the lesion and an ability to effectively illuminate the infected area (20).

Mechanisms of PDT

The mechanism underlying the effects of PDT results from the photons of visible light of an appropriate wavelength interacting with intracellular molecules of the PS (21). Reactive species are produced as a result of the oxidative stress caused by the interaction between the visible light and the biological tissue, and cells are damaged when the reactive oxygen species overwhelm the biochemical defences of the cell (15). A PS is selectively delivered to the target microbial cells and activated by irradiation with light of the appropriate wavelength when taken up by these cells (22). When the PS is activated, type I and/or type II oxidative mechanisms may occur, which underlie the production of free radicals and singlet oxygen, respectively (23). The type I pathway involves electron-transfer reactions from the PS triplet state to a substrate, which results in the production of radical ions that may then react with oxygen to produce cytotoxic species, including superoxide as well as lipid-derived and hydroxyl radicals (24). The type II pathway involves energy transfer from the PS triplet state to ground state molecular oxygen (triplet) to produce excited-state singlet oxygen, which can oxidize various biological molecules, including nucleic acids, proteins and lipids (25,26). These reactive species may then inactivate microbes by damaging cellular components (25), predominantly via the photo-oxidation of nucleic acids, proteins (27) and membrane lipids (28).

The pathway that dominates (either type I or type II) is determined by the general circumstances, including the PS concentration, conditions in the cellular environment, the physicochemical characteristics of the PS and the chemical properties and morphology of the microbial target structures (29). The physiochemical properties of the PS determine its binding affinity to the cell wall of microorganisms; positively charged PS are typically more effective than negative or neutral ones, since, in the majority of cases, the outer surface of microorganisms is negatively charged (30). After the PS binds to the microbial wall, it may either remain outside the microorganism or be translocated to the inner cell membrane in order to induce light- and/or dark-stimulated wall permeability alterations (31). As well as exogenous-acting PS, protoporphyrin IX, which is produced from its precursor 5-aminolevulinic acid (ALA) in the heme biosynthesis signaling pathway, is an endogenous PS that is also important in antimicrobial PDT (32).

PS employed in PDT

A PS, a light source and the presence of significant concentrations of molecular oxygen in the target tissue are all required for PDT (33). The features of an ideal PS include the absence of toxicity, toxic by-products and mutagenic effects, an ability to selectively accumulate in the target tissue, a suitability for topical, oral and intravenous administration, and cost-effectiveness (34).

PS that are used in PDT include chlorines, porphyrins, phenothiazines and phthalocyanines. The phenothiazines used in PDT include orthotoluidine blue and methylene blue (35). Phenothiazines have simple tricyclic planar structures and are cationic compounds. The maximum absorption wavelength is 625 nm for orthotoluidine blue and 656 nm for methylene blue (35). Porphyrins are tetraazamacrocycle compounds that are widely encountered in nature (36). ALA is metabolized to protoporphyrin IX, thus it is not a PS, but rather a porphyrin precursor (2).

Light penetration is also important in PDT (36); light in the blue region penetrates 1.5 mm into the tissue, whereas light in the red region penetrates 3.0 mm. The optimal wavelength to promote photo-killing is ~410 nm (37).

The antifungal action of PDT appears to be strain dependent, and the type of biological medium has been shown to affect the efficacy of PDT in vivo (38). The PS to be used in antifungal PDT must be able to overcome fungal pigments and other substances, as well as the depth of penetration of light into the skin. However, no clinical treatment is currently licensed in the area of antimicrobial PDT (39).

PDT for fungi

The observed effects of PDT on yeasts and dermatophytes have led to the suggestion of its potential use for the treatment of skin mycoses (21). PDT is cost-effective, highly selective and avoids the occurrence of drug resistant strains (40). Therefore, PDT may become a valuable alternative to the already established antifungal drugs if the in vitro and ex vivo results can be transferred to clinical practice (41).

Trichophyton rubrum causes persisting dermatophytosis, and patients with a compromised immune system may suffer from chronic dermatophytosis (42). The ability of the host's defence mechanisms to overcome a dermatophytic infection has been closely associated with the appearance of the infection. The fungal wall is associated with virulence and is also the frequent target of numerous antifungal agents (43). The outermost layer of the fungal wall consists of β-glucan, the second layer contains galactomannans and complex glycoproteins attached to a peptide backbone, and the third layer consists primarily of chitin, which gives the fungal wall its rigidity (44,45). The innermost layer of the wall is the cell membrane (46). Typically, it takes conidia ~2 h to adhere to the skin surface (47). Following the initial attachment to keratinized structures, the conidia germinate and form hyphae, which penetrate the epidermal layer (48). The optimum pH of the proteinases and some of the keratinases produced by the fungi during this initial stage is acidic (49), corresponding to the pH of the skin surface in humans. However, in vitro studies with T. rubrum have shown that the pH of the cultivation medium changes as a function of nutrients used to reach values of pH 8–9 (50). Proteolytic and keratinolytic activity appear to be important virulent factors for dermatophytes.

Typically, the treatment of dermatophytoses involves the administration of an antifungal drug. However, oral antifungal agents may induce side-effects, including hepatotoxicity, and may interact with other drugs (50).

It is important to note that PDT has previously been investigated for the treatment of skin and mucosal infections (50,51). The concentration of the PS in the target tissue and the intensity of photons directed at the target tissue must be considered when evaluating the efficacy of the photodynamic procedure (51). Candida yeasts may cause skin and mucosal infections in patients with local predisposing conditions and are also a major cause of systemic infections, particularly in patients with a compromised immune system (52). Studies have demonstrated that PDT was able to inhibit germ tube and biofilm formation, and reduce adhesion to epithelial buccal cells (53). Dovigo et al (54) reported that biofilms were less susceptible to PDT, as compared with their planktonic counterparts. The effect of PDT has been observed against the dermatophyte T. rubrum (25), and two cases of onychomycosis successfully treated with PDT have previously been described, which involved topical application of an ointment containing 20% ALA.

In vitro studies

The majority of published work on antifungal PDT has centred on in vitro laboratory investigations, involving the use of various fungi, PS and irradiation protocols (55,56). At present, there have been no reports on the development of resistance to antifungal PDT, and the treatment has not been associated with mutagenic effects or genotoxicity. The effects of PDT have predominantly been observed against the dermatophyte T. rubrum (57,58).

In vivo studies

The clinical efficacy of ALA-PDT in the treatment of fungal infections of human skin has previously been investigated (57). Mutagenic effects of photodynamic treatment with chloroaluminum phthalocyanine and RPL068 were not found in Kluyveromyces marxianus (58) nor Candida albicans (59). Following a primary search of 106 articles on databases including, MEDLINE, EMBASE and the Cochrane Library to evaluate the efficacy and safety of PDT for superficial mycoses, it was determined that only seven papers involving 63 patients with superficial mycoses were included. The PS used in all patients was 20% ALA (42).

Adverse effects of ALA-PDT

The overall tolerability of ALA-PDT has been shown to be good, although the adverse effects of ALA-PDT for treating superficial mycoses included a burning sensation during irradiation, erythema, pain, edema and blistering (57).

Limitations and improvement of ALA-PDT

ALA-PDT can be used as a long-term treatment without causing the accumulation of protoporphyrin IX in normal skin (60). ALA is a hydrophilic, zwitterionic molecule with a molecular weight of 167.6 g/mol (61). It is difficult for ALA to penetrate through intact skin (62,63); therefore, improving delivery systems for ALA in the skin will have an important role in the clinical application of ALA-PDT. The enhancement of ALA skin penetration may include physical methods, such as ultrasound, laser, microneedles and iontophoresis, the addition of chemical penetration enhancers, including oleic acid and dimethyl sulfoxide, or the use of lipophilic ALA derivatives or various vehicles to improve the transdermal delivery of ALA (64). Lipophilic ALA ester derivatives may have an enhanced potential for clinical use (65,66). In previous studies, several strategies were used in order to improve ALA penetration into the skin, including iontophoresis (67), lasers (68), microneedles and ultrasound (69).

Mechanisms of penetration enhancers include disruption of the highly ordered structure of stratum corneum lipids, interaction with intercellular proteins and improved partitioning of the drug, co-enhancer, or solvent into the stratum corneum (69). Electron microscopy revealed that a discreet lipid domain is induced within the stratum corneum lipid bilayers upon exposure to oleic acid, which enhanced the permeation of drugs across the skin (54) Friedberg et al (70) reported that oleic acid was able to optimize the skin delivery of ALA in PDT.

The half-life of ALA in the body is ~45 min (70). Vehicles may serve as a solubilization matrix (71). Liposomes, which are microscopic vesicles consisting of one or more membrane-like phospholipid bilayers surrounding an aqueous medium (72,73), are one of the best drug delivery systems for low molecular-weight drugs, such as ALA (74,75).

At present, PDT is used for the prevention and treatment of a variety of malignant skin tumors and inflammatory diseases, including non-melanoma skin cancer, actinic keratoses, acne vulgaris, photorejuvenation, and hidradenitis suppurativa (76,77).

In vitro studies demonstrated that ALA was sufficiently metabolized into protoporphyrin IX and was able to effectively kill T. rubrum and C. albicans (77).

Conclusions

PDT includes the systemic or topical administration of a PS, alongside the selective illumination of a target lesion with light of the appropriate wavelength, in order to cause localized oxidative photodamage and subsequent cell death. Numerous studies have demonstrated that PDT is highly effective in the destruction of fungi in vitro. However, at present, no clinical treatment based on PDT has been licensed. The current study presents in vitro and in vivo and human studies that support antifungal PDT as a new approach against mycoses. In conclusion, antifungal PDT is emerging as an area of interest in the discovery of novel antifungal therapeutic strategies.

References

1 

Wainwright M: Photodynamic antimicrobial chemotherapy (PACT). J Antimicrob Chemother. 42:13–28. 1998. View Article : Google Scholar : PubMed/NCBI

2 

Plaetzer K, Krammer B, Berlanda J, Berr F and Kiesslich T: Photophysics and photochemistry of photodynamic therapy: Fundamental aspects. Lasers Med Sci. 24:259–268. 2009. View Article : Google Scholar : PubMed/NCBI

3 

Bagnato VS, Kurachi C, Ferreira J, Marcassa LG, Sibata CH and Allison RR: PDT experience in Brazil: A regional profile. Photodiagnosis Photodyn Ther. 2:107–118. 2005. View Article : Google Scholar : PubMed/NCBI

4 

Calzavara-Pinton PG, Venturini M and Sala R: A comprehensive overview of photodynamic therapy in the treatment of superficial fungal infections of the skin. J Photochem Photobiol B. 78:1–6. 2005. View Article : Google Scholar : PubMed/NCBI

5 

Yu J, Hsu CH, Huang CC and Chang PY: Development of therapeutic Au-methylene blue nanoparticles for targeted photodynamic therapy of cervical cancer cells. ACS Appl Mater Interfaces. 7:432–441. 2015. View Article : Google Scholar : PubMed/NCBI

6 

de Freitas LM, Soares CP and Fontana CR: Synergistic effect of photodynamic therapy and cisplatin: A novel approach for cervical cancer. J Photochem Photobiol B. 140:365–373. 2014. View Article : Google Scholar : PubMed/NCBI

7 

Gracia-Cazaña T, López MT, Oncins R and Gilaberte Y: Successful treatment of sequential therapy in digital Bowen's disease with methyl aminolevulinate photodynamic therapy and topical diclofenac 3% in hyaluronan 2.5% gel. Dermatol Ther (Heidelb). 28:341–343. 2015. View Article : Google Scholar

8 

Jung SE, Kim SK and Kim YC: Photodynamic therapy in Bowen disease of the first web space of the hand. Ann Dermatol. 27:76–78. 2015. View Article : Google Scholar : PubMed/NCBI

9 

Trushina OI, Novikova EG, Sokolov VV, Filonenko EV, Chissov VI and Vorozhtsov GN: Photodynamic therapy of virus-associated precancer and early stages cancer of cervix uteri. Photodiagn Photodyn Ther. 5:256–259. 2008. View Article : Google Scholar

10 

Cassidy CM, Tunney MM, McCarron PA and Donnelly RF: Drug delivery strategies for photodynamic antimicrobial chemotherapy: From benchtop to clinical practice. J Photochem Photobiol B. 95:71–80. 2009. View Article : Google Scholar : PubMed/NCBI

11 

Wang Q, Yuan D, Liu W, Chen J, Lin X, Cheng S, Li F and Duan X: Use of Optical Fiber Imported Intra-Tissue Photodynamic Therapy for Treatment of Moderate to Severe Acne Vulgaris. Med Sci Monit. 22:362–366. 2016. View Article : Google Scholar : PubMed/NCBI

12 

Al-Qahtani A, Alkahtani S, Kolli B, Tripathi P, Dutta S, Al-Kahtane AA, Jiang XJ, Ng DK and Chang KP: Amino-phthalocyanine-mediated photodynamic inactivation of Leishmania tropica. Antimicrob Agents Chemother: Jan 11. 2016 (Epub ahead of print). AAC: 01879-15. 2016. View Article : Google Scholar

13 

Gupta AK, Einarson TR, Summerbell RC and Shear NH: An overview of topical antifungal therapy in dermatomycoses. A North American perspective. Drugs. 55:645–674. 1998. View Article : Google Scholar : PubMed/NCBI

14 

Calzavara-Pinton PG, Venturini M, Capezzera R, Sala R and Zane C: Photodynamic therapy of interdigital mycoses of the feet with topical application of 5-aminolevulinic acid. Photodermatol Photoimmunol Photomed. 20:144–147. 2004. View Article : Google Scholar : PubMed/NCBI

15 

Henderson BW and Dougherty TJ: How does photodynamic therapy work? Photochem Photobiol. 55:145–157. 1992. View Article : Google Scholar : PubMed/NCBI

16 

Drake LA, Dinehart SM, Farmer ER, Goltz RW, Graham GF, Hordinsky MK, Lewis CW, Pariser DM, Skouge JW, Webster SB, et al: Guidelines/Outcomes Committee. American Academy of Dermatology: Guidelines of care for superficial mycotic infections of the skin: Tinea capitis and tinea barbae. J Am Acad Dermatol. 34:290–294. 1996. View Article : Google Scholar : PubMed/NCBI

17 

Brown GD, Denning DW, Gow NA, Levitz SM, Netea MG and White TC: Hidden killers: Human fungal infections. Sci Transl Med. 4:165rv132012. View Article : Google Scholar : PubMed/NCBI

18 

Cowen LE: The evolution of fungal drug resistance: Modulating the trajectory from genotype to phenotype. Nat Rev Microbiol. 6:187–198. 2008. View Article : Google Scholar : PubMed/NCBI

19 

Soergel P and Hillemanns P: Photodynamic therapy for intraepithelial neoplasia of the lower genital tract. Photodiagn Photodyn Ther. 7:10–14. 2010. View Article : Google Scholar

20 

Smijs TG and Pavel S: The susceptibility of dermatophytes to photodynamic treatment with special focus on Trichophyton rubrum. Photochem Photobiol. 87:2–13. 2011. View Article : Google Scholar : PubMed/NCBI

21 

Hamblin MR and Hasan T: Photodynamic therapy: A new antimicrobial approach to infectious disease? Photochem Photobiol Sci. 3:436–450. 2004. View Article : Google Scholar : PubMed/NCBI

22 

Maisch T: A new strategy to destroy antibiotic resistant microorganisms: Antimicrobial photodynamic treatment. Mini Rev Med Chem. 9:974–983. 2009. View Article : Google Scholar : PubMed/NCBI

23 

Wilson BC and Patterson MS: The physics, biophysics and technology of photodynamic therapy. Phys Med Biol. 53:R61–R109. 2008. View Article : Google Scholar : PubMed/NCBI

24 

Athar M, Mukhtar H and Bickers DR: Differential role of reactive oxygen intermediates in photofrin-I- and photofrin-II-mediated photoenhancement of lipid peroxidation in epidermal microsomal membranes. J Invest Dermatol. 90:652–657. 1988. View Article : Google Scholar : PubMed/NCBI

25 

Redmond RW and Gamlin JN: A compilation of singlet oxygen yields from biologically relevant molecules. Photochem Photobiol. 70:391–475. 1999. View Article : Google Scholar : PubMed/NCBI

26 

Phoenix DA and Harris F: Light activated compounds as antimicrobial agents - patently obvious? Recent Pat Antiinfect Drug Discov. 1:181–199. 2006. View Article : Google Scholar : PubMed/NCBI

27 

Jori G and Coppellotti O: Inactivation of pathogenic microorganisms by photodynamic techniques: Mechanistic aspects and perspective applications. Antiinfect Agents Med Chem. 6:119–131. 2007. View Article : Google Scholar

28 

Smijs TG and Schuitmaker HJ: Photodynamic inactivation of the dermatophyte Trichophyton rubrum. Photochem Photobiol. 77:556–560. 2003. View Article : Google Scholar : PubMed/NCBI

29 

Girotti AW and Kriska T: Role of lipid hydroperoxides in photo-oxidative stress signaling. Antioxid Redox Signal. 6:301–310. 2004. View Article : Google Scholar : PubMed/NCBI

30 

Bertoloni G, Zambotto F, Conventi L, Reddi E and Jori G: Role of specific cellular targets in the hematoporphyrin-sensitized photoinactivation of microbial cells. Photochem Photobiol. 46:695–698. 1987. View Article : Google Scholar : PubMed/NCBI

31 

Merchat M, Bertolini G, Giacomini P, Villanueva A and Jori G: Meso-substituted cationic porphyrins as efficient photosensitizers of gram-positive and gram-negative bacteria. J Photochem Photobiol B. 32:153–157. 1996. View Article : Google Scholar : PubMed/NCBI

32 

Lee JW, Kim BJ and Kim MN: Photodynamic therapy: New treatment for recalcitrant Malassezia folliculitis. Lasers Surg Med. 42:192–196. 2010. View Article : Google Scholar : PubMed/NCBI

33 

Allison RR, Downie GH, Cuenca R, Hu XH, Childs CJ and Sibata CH: Photosensitizers in clinical PDT. Photodiagn Photodyn Ther. 1:27–42. 2004. View Article : Google Scholar

34 

Magalhães JL, Moreira LM, Rodrigues-Filho UP, Giz MJ, Pereira-da-Silva MA, Landers R, Vinhas RCG and Nascente PAP: Surface chemistry of iron tetraazamacrocycle on the aminopropyl-modified surface of oxidized n-Si(100) by AFM and XPS. Surf Interface Anal. 33:293–298. 2002. View Article : Google Scholar

35 

Moreira LM, dos Santos FV, Lyon JP, Maftoum-Costa M, Pacheco-Soares C and da Silva NA: Photodynamic therapy: Porphyrins and phthalocyanines as photosensitizers. J Chem. 61:741–754. 2008.

36 

Moreira LM, Ribelatto JC and Imasato H: Ruffled and planar conformations of the porphyrin ring in complexes and heme proteins: Physical-chemistry properties and spectroscopic implications. Quim Nova. 27:958–963. 2004.(In Portuguese). View Article : Google Scholar

37 

Nyman ES and Hynninen PH: Research advances in the use of tetrapyrrolic photosensitizers for photodynamic therapy. J Photochem Photobiol B. 73:1–28. 2004. View Article : Google Scholar : PubMed/NCBI

38 

Plaetzer K, Krammer B, Berlanda J, Berr F and Kiesslich T: Photophysics and photochemistry of photodynamic therapy: Fundamental aspects. Lasers Med Sci. 24:259–268. 2009. View Article : Google Scholar : PubMed/NCBI

39 

Calzavara-Pinton P, Rossi MT, Sala R and Venturini M: Photodynamic antifungal chemotherapy. Photochem Photobiol. 88:512–522. 2012. View Article : Google Scholar : PubMed/NCBI

40 

Maisch T, Szeimies RM, Lehn N and Abels C: Antibacterial photodynamic therapy. A new treatment for superficial bacterial infections? Hautarzt. 56:1048–1055. 2005.(In German). PubMed/NCBI

41 

Ragàs X, Agut M and Nonell S: Singlet oxygen in Escherichia coli: New insights for antimicrobial photodynamic therapy. Free Radic Biol Med. 49:770–776. 2010. View Article : Google Scholar : PubMed/NCBI

42 

Odom R: Dermatologic manifestations of AIDS. J Am Podiatr Med Assoc. 78:127–129. 1988. View Article : Google Scholar : PubMed/NCBI

43 

Martinez-Rossi NM, Peres NT and Rossi A: Antifungal resistance mechanisms in dermatophytes. Mycopathologia. 166:369–383. 2008. View Article : Google Scholar : PubMed/NCBI

44 

Grappel SF, Bishop CT and Blank F: Immunology of dermatophytes and dermatophytosis. Bacteriol Rev. 38:222–250. 1974.PubMed/NCBI

45 

San-Blas G: The cell wall of fungal human pathogens: Its possible role in host-parasite relationships. Mycopathologia. 79:159–184. 1982. View Article : Google Scholar : PubMed/NCBI

46 

Deacon JW: The moulds of man. Fungal Biology. Blackwell Publishing Ltd. 322–338. 2006.

47 

Zurita J and Hay RJ: Adherence of dermatophyte microconidia and arthroconidia to human keratinocytes in vitro. J Invest Dermatol. 89:529–534. 1987. View Article : Google Scholar : PubMed/NCBI

48 

Apodaca G and McKerrow JH: Regulation of Trichophyton rubrum proteolytic activity. Infect Immun. 57:3081–3090. 1989.PubMed/NCBI

49 

Brasch J and Zaldua M: Enzyme patterns of dermatophytes. Mycoses. 37:11–16. 1994. View Article : Google Scholar : PubMed/NCBI

50 

Maranhão FC, Paião FG and Martinez-Rossi NM: Isolation of transcripts over-expressed in human pathogen Trichophyton rubrum during growth in keratin. Microb Pathog. 43:166–172. 2007. View Article : Google Scholar : PubMed/NCBI

51 

Gupta AK and Cooper EA: Update in antifungal therapy of dermatophytosis. Mycopathologia. 166:353–367. 2008. View Article : Google Scholar : PubMed/NCBI

52 

Lyon JP, Costa SC, Totti VMG, Munhoz MF and de Resende MA: Predisposing conditions for Candida spp. carriage in the oral cavity of denture wearers and individuals with natural teeth. Can J Microbiol. 52:462–467. 2006. View Article : Google Scholar : PubMed/NCBI

53 

Munin E, Giroldo LM, Alves LP and Costa MS: Study of germ tube formation by Candida albicans after photodynamic antimicrobial chemotherapy (PACT). J Photochem Photobiol B. 88:16–20. 2007. View Article : Google Scholar : PubMed/NCBI

54 

Dovigo LN, Pavarina AC, Mima EG, Giampaolo ET, Vergani CE and Bagnato VS: Fungicidal effect of photodynamic therapy against fluconazole-resistant Candida albicans and Candida glabrata. Mycoses. 54:123–130. 2011. View Article : Google Scholar : PubMed/NCBI

55 

Fuchs BB, Tegos GP, Hamblin MR and Mylonakis E: Susceptibility of Cryptococcus neoformans to photodynamic inactivation is associated with cell wall integrity. Antimicrob Agents Chemother. 51:2929–2936. 2007. View Article : Google Scholar : PubMed/NCBI

56 

Prates RA, Kato IT, Ribeiro MS, Tegos GP and Hamblin MR: Influence of multidrug efflux systems on methylene blue-mediated photodynamic inactivation of Candida albicans. J Antimicrob Chemother. 66:1525–1532. 2011. View Article : Google Scholar : PubMed/NCBI

57 

Calzavara-Pinton PG, Venturini M, Capezzera R, Sala R and Zane C: Photodynamic therapy of interdigital mycoses of the feet with topical application of 5-aminolevulinic acid. Photodermatol Photoimmunol Photomed. 20:144–147. 2004. View Article : Google Scholar : PubMed/NCBI

58 

Smijs TGM, Bouwstra JA, Schuitmaker HJ, Talebi M and Pavel S: A novel ex vivo skin model to study the susceptibility of the dermatomycete Trichophyton rubrum to photodynamic treatment in different growth phases. J Antimicrob Chemother. 59:433–440. 2007. View Article : Google Scholar : PubMed/NCBI

59 

Peres NT, Sanches PR, Falcão JP, Silveira HC, Paião FG, Maranhão FC, Gras DE, Segato F, Cazzaniga RA, Mazucato M, et al: Transcriptional profiling reveals the expression of novel genes in response to various stimuli in the human dermatophyte Trichophyton rubrum. BMC Microbiol. 10:392010. View Article : Google Scholar : PubMed/NCBI

60 

Akilov OE, Kosaka S, O'Riordan K and Hasan T: Parasiticidal effect of delta-aminolevulinic acid-based photodynamic therapy for cutaneous leishmaniasis is indirect and mediated through the killing of the host cells. Exp Dermatol. 16:651–60. 2007. View Article : Google Scholar : PubMed/NCBI

61 

Wiegell SR, Stender IM, Na R and Wulf HC: Pain associated with photodynamic therapy using 5-aminolevulinic acid or 5-aminolevulinic acid methylester on tape-stripped normal skin. Arch Dermatol. 139:1173–1177. 2003. View Article : Google Scholar : PubMed/NCBI

62 

Sil S, Bose T, Roy D and Chakraborti AS: Protoporphyrin IX-induced structural and functional changes in human red blood cells, haemoglobin and myoglobin. J Biosci. 29:281–291. 2004. View Article : Google Scholar : PubMed/NCBI

63 

Di Venosa G, Hermida L, Fukuda H, Defain MV, Rodriguez L, Mamone L, MacRobert A, Casas A and Batlle A: Comparation of liposomal formulations of ALA Undecanoyl ester for its use in photodynamic therapy. J Photochem Photobiol B. 96:152–158. 2009. View Article : Google Scholar : PubMed/NCBI

64 

Morrow DI, McCarron PA, Woolfson AD, Juzenas P, Juzeniene A, Iani V, Moan J and Donnelly RF: Hexyl aminolaevulinate is a more effective topical photosensitiser precursor than methyl aminolaevulinate and 5-aminolaevulinic acids when applied in equimolar doses. J Pharm Sci. 99:3486–3498. 2010. View Article : Google Scholar : PubMed/NCBI

65 

van den Akker JT, Iani V, Star WM, Sterenborg HJ and Moan J: Topical application of 5-aminolevulinic acid hexyl ester and 5-aminolevulinic acid to normal nude mouse skin: Differences in protoporphyrin IX fluorescence kinetics and the role of the stratum corneum. Photochem Photobiol. 72:681–689. 2000. View Article : Google Scholar : PubMed/NCBI

66 

Gaullier JM, Berg K, Peng Q, Anholt H, Selbo PK, Ma LW and Moan J: Use of 5-aminolevulinic acid esters to improve photodynamic therapy on cells in culture. Cancer Res. 57:1481–1486. 1997.PubMed/NCBI

67 

Lopez RF, Bentley MV, Begoña Delgado-Charro M and Guy RH: Optimization of aminolevulinic acid delivery by iontophoresis. J Control Release. 88:65–70. 2003. View Article : Google Scholar : PubMed/NCBI

68 

Kacerovska D, Pizinger K, Kumpova M and Cetkovska P: Genital warts treated by photodynamic therapy. Skinmed. 6:295–297. 2007. View Article : Google Scholar : PubMed/NCBI

69 

Mikolajewska P, Donnelly RF, Garland MJ, Morrow DI, Singh TR, Iani V, Moan J and Juzeniene A: Microneedle pre-treatment of human skin improves 5-aminolevulininc acid (ALA)- and 5-aminolevulinic acid methyl ester (MAL)-induced PpIX production for topical photodynamic therapy without increase in pain or erythema. Pharm Res. 27:2213–2220. 2010. View Article : Google Scholar : PubMed/NCBI

70 

Friedberg JS, Skema C, Baum ED, Burdick J, Vinogradov SA, Wilson DF, Horan AD and Nachamkin I: In vitro effects of photodynamic therapy on Aspergillus fumigatus. J Antimicrob Chemother. 48:105–107. 2001. View Article : Google Scholar : PubMed/NCBI

71 

Fang JY, Hong CT, Chiu WT and Wang YY: Effect of liposomes and niosomes on skin permeation of enoxacin. Int J Pharm. 219:61–72. 2001. View Article : Google Scholar : PubMed/NCBI

72 

Allison RR, Downie GH, Cuenca R, et al: Photosensitizers in clinical PDT. Photodiagnosis Photodyn Ther. 1:27–42. 2004. View Article : Google Scholar : PubMed/NCBI

73 

Kamp H, Tietz HJ, Lutz M, Piazena H, Sowyrda P, Lademann J and Blume-Peytavi U: Antifungal effect of 5-aminolevulinic acid PDT in Trichophyton rubrum. Mycoses. 48:101–107. 2005. View Article : Google Scholar : PubMed/NCBI

74 

Zucker D, Marcus D, Barenholz Y and Goldblum A: Liposome drugs' loading efficiency: A working model based on loading conditions and drug's physicochemical properties. J Control Release. 139:73–80. 2009. View Article : Google Scholar : PubMed/NCBI

75 

Fang JY: Nano- or submicron-sized liposomes as carriers for drug delivery. Chang Gung Med J. 29:358–362. 2006.PubMed/NCBI

76 

Huang L, Huang YY, Mroz P, Tegos GP, Zhiyentayev T, Sharma SK, Lu Z, Balasubramanian T, Krayer M, Ruzié C, et al: Stable synthetic cationic bacteriochlorins as selective antimicrobial photosensitizers. Antimicrob Agents Chemother. 54:3834–3841. 2010. View Article : Google Scholar : PubMed/NCBI

77 

Qiao J, Li R, Ding Y and Fang H: Photodynamic therapy in the treatment of superficial mycoses: An evidence-based evaluation. Mycopathologia. 170:339–343. 2010. View Article : Google Scholar : PubMed/NCBI

Related Articles

Journal Cover

July-2016
Volume 12 Issue 1

Print ISSN: 1792-0981
Online ISSN:1792-1015

Sign up for eToc alerts

Recommend to Library

Copy and paste a formatted citation
x
Spandidos Publications style
Liang Y, Lu LM, Chen Y and Lin YK: Photodynamic therapy as an antifungal treatment (Review). Exp Ther Med 12: 23-27, 2016
APA
Liang, Y., Lu, L., Chen, Y., & Lin, Y. (2016). Photodynamic therapy as an antifungal treatment (Review). Experimental and Therapeutic Medicine, 12, 23-27. https://doi.org/10.3892/etm.2016.3336
MLA
Liang, Y., Lu, L., Chen, Y., Lin, Y."Photodynamic therapy as an antifungal treatment (Review)". Experimental and Therapeutic Medicine 12.1 (2016): 23-27.
Chicago
Liang, Y., Lu, L., Chen, Y., Lin, Y."Photodynamic therapy as an antifungal treatment (Review)". Experimental and Therapeutic Medicine 12, no. 1 (2016): 23-27. https://doi.org/10.3892/etm.2016.3336