Open Access
13 November 2012 Photodynamic therapy in dermatology: past, present, and future
Razvigor Darlenski, Joachim W. Fluhr
Author Affiliations +
Abstract
Photodynamic therapy (PDT) is a noninvasive therapeutic method first introduced in the field of dermatology. It is mainly used for the treatment of precancerous and superficial malignant skin tumors. Today PDT finds new applications not only for nononcologic dermatoses but also in the field of other medical specialties such as otorhinolaryngology, ophthalmology, neurology, gastroenterology, and urology. We are witnessing a broadening of the spectrum of skin diseases that are treated by PDT. Since its introduction, PDT protocol has evolved significantly in terms of increasing method efficacy and patient safety. In this era of evidence-based medicine, it is expected that much effort will be put into creating a worldwide accepted consensus on PDT. A review on the current knowledge of PDT is given, and the historical basis of the method's evolution since its introduction in the 1900s is presented. At the end, future challenges of PDT are focused on discussing gaps that exist for research in the field.

1.

Introduction

In the past decades, photodynamic therapy (PDT) has gained wide popularity in medicine and in dermatology in particular. Since its introduction, the procedure has evolved in terms of increasing safety and efficacy. Today PDT is used worldwide not only in the field of dermatology but also for adjuvant treatment in lung, brain, esophageal, biliary and urinary tract cancer.1 In dermatology, this method is mainly used as a primary treatment for malignant and premalignant skin lesions, while many other nononcological applications have emerged due to the efforts of different study groups.

1.1.

Principle of PDT

PDT is based on the photodynamic reaction: use of a light-sensitive substance (a photosensitizer), combined with light of a visible wavelength, to destroy target cells. This toxic biochemical reaction is oxygen-mediated. The photosensitizer absorbs a photon of visible light and then transfers most of the absorbed energy to a molecule of oxygen (Fig. 1). This converts it into a relatively strong oxidizing agent known as singlet oxygen. As a consequence, in the tissues that have accumulated the sensitizer, light-induced singlet oxygen exerts a cytotoxic effect by causing lethal oxidative damage to biologically important structures.

Fig. 1

Mechanism of the photodynamic reaction: the photosensitizer (S) is activated by the visible light. Most absorbed energy is transferred to a molecule of oxygen, which in turn is transformed into highly reactive and cytotoxic singlet oxygen (O21). The oxidative damage is limited to the tissues rich in S.

JBO_18_6_061208_f001.png

The selection of a proper photosensitizer has posed the greatest challenge in the years of PDT development. A substance that is naturally occurring offers a sufficient balance between selective tissue accumulation and relatively short clearance of the body, namely the protoporphyrin IX (PpIX). PpIX is a natural photosensitizer that can be made by the human body and is an intermediate product in the biosynthesis of heme (Fig. 2). It accumulates in rapidly proliferating cells of premalignant and malignant lesions, as well as in other structures, such as blood vessels, melanin, and sebaceous glands. In addition, malignant cells exert reduced ferrochetalase activity, resulting in excessive accumulation of intracellular PpIX.2

Fig. 2

Biosynthesis pathway of heme.

JBO_18_6_061208_f002.png

In PDT, aminolevulinic acid (ALA) or its methylated derivative—methyl-aminolevulinate (MAL)—is applied to the skin for varying periods of time, thus bypassing the rate-limiting step in the biosynthesis of heme. This leads to the conversion of ALA/MAL to PpIX. The activation of the sensitizer is accomplished by light with a specific wavelength that corresponds to the maximum absorption spectra of the sensitizer. In an ideal situation, the consecutive tissue damage is selective and only the rapidly proliferating tissue with accumulated PpIX will be destroyed with any surrounding tissue damage.

2.

Historical Perspective

It was 1900 when Raab first reported the destruction of the Paramecium caudatum cells by exposure to combined acridine orange and light.3 In contrast, neither the dye nor the light separately induced the cellular death. In the next decade, the extensive work of von Tappeiner contributed to the development of the concept of PDT. He first studied the photodynamic effect in protozoa by applying aniline dyes and fluorescent light.4 One year later he described the first cases of PDT in humans by using eosin as a photosensitizer to treat a number of conditions such as condylomata lata, lupus vulgaris, and nonmelanoma skin cancer (NMSC).5 In the later years, different photosensitizers have been introduced, and hematoporphyrin is probably the most widely studied. However, the clearance of the substance from the tissue was very slow and the phototoxic reaction persisted for a long period of time.

In the late 1970s, a new substance was introduced and recently has become a gold standard in the PDT, namely hematoporphyrin purified derivative.6 The photoactivation was performed by visible red light, but again the accumulation in the skin lasted for up to several months. It was Kennedy in 1990 who first used ALA for topical PDT on the skin.7 Due to the low molecular size, ALA easily penetrated the stratum corneum. In addition, it was cleared far more rapidly than the formerly used sensitizers and phototoxicity was observed only several days after the ALA application.7

3.

Indications for PDT in Dermatology

Since its introduction, the list of PDT applications has consistently grown. Indisputably, actinic keratoses and NMSC have been the most widely used, so they will be the focus of this paper. A list of the current PDT applications is provided in Table 1.

Table 1

Applications of PDT in dermatology.

Malignant and premalignant conditionsNononcologic skin diseases
Aktinic keratoses (and associated photodamage)Acne
Actinic chelitisPsoriasis vulgaris
Superficial basal cell carcinomaMolluscum contagiosum
Superficial squamous cell carcinomaHuman papillomavirus infection
Field cancerization of the skinHerpes virus infection
Bowen’s diseaseErythrasma
Mammary and extra-mammary Paget’s diseaseAlopecia areata
Erythroplasia of QueyratHirsutism
Cutaneous T-cell lymphomaSebaceous gland hyperplasia
Kaposi’s sarcomaNaevus sebaceus
Malignant melanomaHidradenitis suppurativa
KeratoacanthomaKeloids and hypertrophic scars
Gorlin syndrome (multiple nevoid basasl cell carcinoma)Pigmented purpuric dermatosis
Penile and vulvar intraepithelial neoplasiaDisseminated actinic porokeratosis
Langerhans cell histiocytosisErosive pustular dermatosis of the scalp
Skin metastasesAcquired perforating dermatosis
Cutaneous sarcoidosis
Cutaneous leishmaniasis
Lichen planus
Morphea
Darier’s disease (diskeratosis follicularis)
Lichen sclerosus et atrophicus
Lymphocytic infiltration of the skin
Pseudepitheliomatous hyperplasia
Skin and nail mycoses
Acinetobacter baumannii skin infections
Wound healing
Photorejuvenation
Permanent depilation

Beyond therapeutic indications, the selective accumulation of the photosensitizer is used in the so-called fluorescent diagnostics. In this setting, the skin area of interest is illuminated by ultraviolet light (most often by using a Wood lamp) which allows the visualization of the accumulated sensitizer in the skin. The method is used in preoperative planning for the exact delineation of the tumor borders as well for control of anti-cancer therapies.8,9

4.

Factors in PDT

4.1.

Sensitizer

Although the substances (ALA and MAL) used for local PDT in dermatology are generally referred to as photosensitizers, they are prodrugs. Once delivered to the viable epidermis, ALA/MAL is converted to PpIX, which is the endogenous photactivating agent. Within the next 24 to 48 h, PpIX is transformed to the photodynamically inactive heme.10

δ-5-ALA is a low molecular weight, hydrophilic molecule that can penetrate the stratum corneum and then can be included in the biosynthesis pathway of heme [Fig. 3(a)]. In the United States, ALA is marketed as 20% topical solution of hydrochloride salt. A variety of custom-made preparations as emulsions and gels are available in practice.

Fig. 3

Chemical formula of aminolevulinic acid (a) and methylaminolevulinate (b).

JBO_18_6_061208_f003.png

In addition, further substances can enhance the accumulation of PpIX, such as desferrioxamine, and the adding of DMSO and EDTA to ALA can enhance the penetration of the precursor.11

Esters of ALA are more commonly used in Europe. They are lipophilic derivates of ALA, which allows enhanced penetration through the lipid bilayers of the horny layer.12 It has been shown that MAL [Fig. 3(b)] possesses better tumor selectivity and less patient discomfort compared to ALA.13 Sixteen percent MAL cream is registered in both the United States and Europe. During photodynamic diagnosis, MAL provided higher tumor contrast than ALA in basal cell carcinoma visualization.14 The authors concluded that MAL should be preferred for use in fluorescence diagnostics.

In recent years, new systems to carry sensitizers to the cells have been developed, such as nanostructural materials, polymeric and liposomal formulations of the sensitizers, and lipid nano-carrier-mediated nuclear targeting carriers.15

The list of substances applied in PDT is increasing and includes chlorins, bacteriochlorins, auxins, pheophorbides, purpurins, phthalocyanines, and naphthalocyanines.16,17

4.2.

Light Source

Different light sources of coherent (lasers) or incoherent origin are applied in PDT. Coherence is lost within the first millimeters of penetration into the skin;18 therefore, the use of such light sources is not an obligatory prerequisite. Furthermore the use of lasers is more expensive and can be related to some difficulties during the exploitation.19 Incoherent light sources remain the golden standard for PDT, including a variety of broadband lamps, light-emitting diodes, and intense pulsed light systems. Table 2 summarizes the reports in the literature data about the light intensity and dosing in PDT with incoherent light sources. Beyond blue and red light, green and white light sources have also been occasionally reported in PDT.19

Table 2

Light dose and intensity for incoherent light sources for PDT.

Light sourceIndicationsDoseIntensity
Broad spectrum red lightOncologic diseases100150J/cm2100200mW/cm2
Broad spectrum red lightInflammatory dermatoses1040J/cm25070mW/cm2
Light emitting diodes Oncologic diseases3750J/cm2Up to 200mW/cm2

Porphyrins exhibit peak absorption at approximately 405 nm (Soret band; blue light spectrum) as well as several Q bands with absorption peaks in the red light spectrum. Red light exhibits deeper penetration profiles in the skin, therefore it is the only light approved for PDT of skin tumors.16

A recently developed protocol proposed the so-called daylight-mediated PDT for actinic keratoses.20 In this setting, MAL is applied on the entire affected skin field and the patients are exposed to daylight with no further illumination with artificial light sources. A randomized multicenter study showed that this method is efficient even after a single treatment session.20 A natural daylight exposure of an hour and a half was sufficient to gain efficacy. Thin lesions responded better than the moderate and thick actinic keratoses.

5.

Adverse Events in PDT

PDT is generally well tolerated. The most common adverse events include pain and a burning sensation limited to the term of the irradiation and several hours afterwards. Larger irradiation areas and sites with rich innervation, e.g., the head, hands, and perineum, are associated with greater pain sensation.13 A correlation between pain and the dose/intensity of the used light was also evidenced.13,21 Pain is greater in a second session compared with the first as shown by a single study.22 This could potentially cause a decrease in the patient’s compliance. Different strategies for decreasing the pain in PDT have been proposed. Table 3 summarizes the pain management strategies.

Table 3

Pain management strategies in PDT.

MethodDescriptionEfficacyLimitation
Pre-irradiation systemic analgesiaIntake of analgesics such as metamizole, piritramide, or benzodiazepines, before the session Limited efficacy; sometimes reanimation unit requiredPossible drug interactions
General anesthesiaShort-term systemic anesthetic applicationGoodAnesthetist required
Cool air Ventilation during irradiationDecreases pain, but does not eliminate it completelySpecial equipment required
Topical anestheticsApplication of creams or gels with topical anestheticsLimited reduction of painNot advised as the anesthetic might interfere with the acidity of the ALA/MAL-preparation
Injectable topical anestheticsInfiltration anesthesia and nerve blockEffectiveTo be used pure, without vasoconstrictive agents in the preparation
Interrupting the sessionFor intervals of 3 min; spraying cold water spray in the intervalReduction of painProlongation of the session; no data on the long-term efficacy
Thermal water spraySprays used from the 4th hour after the sessionReduction of pain form day 3 to day 6 after PDTNo confirmatory studies; Does not deal with the pain during the irradiation session

Further local adverse events include erythema, edema, erosions, aseptic pustulosis, necrosis of the tumor, scarring, hyper- and hypopigmentation, and loss of hair. Several cases of contact allergic dermatitis to ALA23 and MAL24 have been described. Two reports describe possible coincidental association of PDT with carcinogenicity.25 Experimental studies in mice showed that repetitive treatments with ALA-PDT even delay photo-induced carcinogenesis.26

6.

Future of PDT

In the past century, PDT has been established as a safe, efficacious, and generally well-tolerated therapeutic method in dermatology. Today, devices for performing PDT in ambulatory settings are available. In our view, the five-year perspectives for PDT can be summarized into the following fields:

  • - Novel sensitizer development and new carrier systems to the skin, e.g., nanotechnologies: We are witnessing the constant development of new molecules and delivery systems. The challenge in this field would be a faster and more selective tissue accumulation of the sensitizer, as well as the shortened clearance period.

  • - New light sources: A step forward in this direction is the implementation of light-emitting-diode technologies in PDT. Decreasing the intensity of the light, and thus the subjective discomfort, in parallel to keeping the therapeutic efficacy, poses a challenge to researchers.

  • - Reduction of pain during and after treatment sessions: New physical and/or chemical (medicamentous) methods should be investigated as the major adverse event during PDT is the pain sensation. These should not interfere with the PDT procedure and pharmacokinetics of the sensitizers in the skin.

  • - Standardization of PDT procedures worldwide: Efforts in this area have been made and certain international consensus and guidelines for PDT already exist.27 One of the major roles of such a consensus document exerts protective effects over medical practitioners as a part of the evidence-based medicine.

  • - New indications for PDT: This is an area which is constantly enriched by the multiple reports for the successive application to a variety of skin diseases. PDT has already been successfully applied in the treatment of skin infections with multi-drug-resistant microorganisms such as MRSA.28

The constant and dynamic development of novelties in the field is a certain guarantee for the future of PDT in dermatology.

References

1. 

P. Babilaset al., “Photodynamic therapy in dermatology: state-of-the-art,” Photodermatol. Photoimmunol. Photomed., 26 (3), 118 –132 (2010). http://dx.doi.org/10.1111/(ISSN)1600-0781 PPPHEW 0905-4383 Google Scholar

2. 

D. Toumaet al., “A trial of short incubation, broad-area photodynamic therapy for facial actinic keratoses and diffuse photodamage,” Arch. Dermatol., 140 (1), 33 –40 (2004). http://dx.doi.org/10.1001/archderm.140.1.33 ARDEAC 0003-987X Google Scholar

3. 

O. Raab, “Ueber die wirkung fluoreszierenden stoffe auf infusorien,” Z. Biol., 39 524 –526 (1900). Google Scholar

4. 

H. V. TappeinerA. Jodblauer, “Uber die wirkung der photodynamischen (fluorescierenden) staffe auf protozoan und enzyme,” Dtsch. Arch. Klin. Med., 80 427 –487 (1904). Google Scholar

5. 

A. JesionekH. V. Tappeiner, “Behandlung der hautcarcinome nut fluorescierenden stoffen,” Dtsch. Arch. Klin. Med., 85 223 –227 (1905). Google Scholar

6. 

T. J. Doughertyet al., “Photoradiation therapy for the treatment of malignant tumors,” Cancer Res., 38 (8), 2628 –2635 (1978). CNREA8 0008-5472 Google Scholar

7. 

J. C. KennedyR. H. PottierD. C. Pross, “Photodynamic therapy with endogenous protoporphyrin IX: basic principles and present clinical experience,” J. Photochem. Photobiol. B, 6 (1–2), 143 –148 (1990). http://dx.doi.org/10.1016/1011-1344(90)85083-9 JPPBEG 1011-1344 Google Scholar

8. 

E. TierneyJ. PetersenC. W. Hanke, “Photodynamic diagnosis of tumor margins using methyl aminolevulinate before Mohs micrographic surgery,” J. Am. Acad. Dermatol., 64 (5), 911 –918 (2011). http://dx.doi.org/10.1016/j.jaad.2010.03.045 JAADDB 0190-9622 Google Scholar

9. 

J. TyrrellS. CampbellA. Curnow, “Validation of a non-invasive fluorescence imaging system to monitor dermatological PDT,” Photodiagn. Photodyn. Ther., 7 (2), 86 –97 (2010). http://dx.doi.org/10.1016/j.pdpdt.2010.03.002 1572-1000 Google Scholar

10. 

E. S. MarmurC. D. SchmultsD. J. Goldberg, “A review of laser and photodynamic therapy for the treatment of nonmelanoma skin cancer,” Dermatol. Surg., 30 (Pt. 2), 264 –271 (2004). http://dx.doi.org/10.1111/dsu.2004.30.issue-s2 1076-0512 Google Scholar

11. 

K. KalkaH. MerkH. Mukhtar, “Photodynamic therapy in dermatology,” J. Am. Acad. Dermatol., 42 (3), 389 –413 (2000). http://dx.doi.org/10.1016/S0190-9622(00)90209-3 JAADDB 0190-9622 Google Scholar

12. 

N. Fotinoset al., “5-Aminolevulinic acid derivatives in photomedicine: Characteristics, application and perspectives,” Photochem. Photobiol., 82 (4), 994 –1015 (2006). http://dx.doi.org/10.1562/2006-02-03-IR-794 PHCBAP 0031-8655 Google Scholar

13. 

Y. N. Chaveset al., “Pain in photodynamic therapy: mechanism of action and management strategies,” An. Bras. Dermatol., 87 (4), 521 –529 (2012). http://dx.doi.org/10.1590/S0365-05962012000400001 0365-0596 Google Scholar

14. 

C. Sandberget al., “Fluorescence diagnostics of basal cell carcinomas comparing methyl-aminolaevulinate and aminolaevulinic acid and correlation with visual clinical tumor size,” Acta Derm. Venereol., 91 (4), 398 –403 (2011). http://dx.doi.org/10.2340/00015555-1068 0001-5555 Google Scholar

15. 

M. KepczynskiM. DzieciuchM. Nowakowska, “Nanostructural hybrid sensitizers for photodynamic therapy,” Curr. Pharm. Des., 18 (18), 2607 –2621 (2012). http://dx.doi.org/10.2174/138161212800492877 1381-6128 Google Scholar

16. 

C. A. Mortonet al., “Guidelines for topical photodynamic therapy: report of a workshop of the British Photodermatology Group,” Br. J. Dermatol., 146 (4), 552 –567 (2002). http://dx.doi.org/10.1046/j.1365-2133.2002.04719.x BJDEAZ 1365-2133 Google Scholar

17. 

S. Y. Huhet al., “The effect of photodynamic therapy using indole-3-acetic acid and green light on acne vulgaris,” Ann. Dermatol., 24 (1), 56 –60 (2012). http://dx.doi.org/10.5021/ad.2012.24.1.56 ANDEEM Google Scholar

18. 

R. M. Szeimieset al., “A possible new incoherent lamp for photodynamic treatment of superficial skin lesions,” Acta Derm. Venereol., 74 (2), 117 –119 (1994). ADVEA4 0001-5555 Google Scholar

19. 

P. BabilasM. LandthalerR. M. Szeimies, “Photodynamic therapy in dermatology,” Eur. J. Dermatol., 16 (4), 340 –348 (2006). http://dx.doi.org/10.1016/S0190-9622(00)90209-3 EJDEE4 1167-1122 Google Scholar

20. 

S. R. Wiegellet al., “Daylight-mediated photodynamic therapy of moderate to thick actinic keratoses of the face and scalp: a randomized multicentre study,” Br. J. Dermatol., 166 (6), 1327 –1332 (2012). http://dx.doi.org/10.1111/bjd.2012.166.issue-6 BJDEAZ 1365-2133 Google Scholar

21. 

P. Babilaset al., “Variable pulsed light is less painful than light-emitting diodes for topical photodynamic therapy of actinic keratosis: a prospective randomized controlled trial,” Br. J. Dermatol., 157 (1), 111 –117 (2007). http://dx.doi.org/10.1111/bjd.2007.157.issue-1 BJDEAZ 1365-2133 Google Scholar

22. 

D. R. Leffet al., “The effect of local cooling on pain perception during infiltration of local anesthetic agents, a prospective randomized controlled trial,” Anesthesia, 62 (7), 677 –682 (2007). http://dx.doi.org/10.1111/ana.2007.62.issue-7 0003-2409 Google Scholar

23. 

H. C. WulfP. Philipsen, “Allergic contact dermatitis to 5-aminolaevulinic acid methylester but not to 5-aminolaevulinic acid after photodynamic therapy,” Br. J. Dermatol., 150 (1), 143 –145 (2004). http://dx.doi.org/10.1111/bjd.2004.150.issue-1 BJDEAZ 1365-2133 Google Scholar

24. 

J. M. Jungerstedet al., “Allergic reactions to Metvix (ALA-ME),” Contact Dermatitis., 58 (3), 184 –186 (2008). http://dx.doi.org/10.1111/cod.2008.58.issue-3 CODEDG 0105-1873 Google Scholar

25. 

P. BabilasR. M. Szeimies, “The use of photodynamic therapy in dermatology,” G. Ital. Dermatol. Venereol., 145 (5), 613 –630 (2010). GIDVDZ 0392-0488 Google Scholar

26. 

I. M. Stenderet al., “Photodynamic therapy with topical delta-aminolevulinic acid delays UV photocarcinogenesis in hairless mice,” Photochem. Photobiol., 66 (4), 493 –496 (1997). http://dx.doi.org/10.1111/php.1997.66.issue-4 PHCBAP 0031-8655 Google Scholar

27. 

L. R. Braathenet al., “Photodynamic therapy for skin field cancerization: an international consensus. International Society for Photodynamic Therapy in Dermatology,” J. Eur. Acad. Dermatol. Venereol., 26 (9), 1063 –1066 (2012). http://dx.doi.org/10.1111/jdv.2012.26.issue-9 JEAVEQ 0926-9959 Google Scholar

28. 

N. KashefG. R. Sharif AbadiG. E. Djavid, “Phototoxicity of phenothiazinium dyes against methicillin-resistant Staphylococcus aureus and multi-drug resistant Escherichia coli,” Photodiagn. Photodyn Ther., 9 (1), 11 –15 (2012). http://dx.doi.org/10.1016/j.pdpdt.2011.11.004 1572-1000 Google Scholar
© 2012 Society of Photo-Optical Instrumentation Engineers (SPIE) 0091-3286/2012/$25.00 © 2012 SPIE
Razvigor Darlenski and Joachim W. Fluhr "Photodynamic therapy in dermatology: past, present, and future," Journal of Biomedical Optics 18(6), 061208 (13 November 2012). https://doi.org/10.1117/1.JBO.18.6.061208
Published: 13 November 2012
Lens.org Logo
CITATIONS
Cited by 59 scholarly publications.
Advertisement
Advertisement
RIGHTS & PERMISSIONS
Get copyright permission  Get copyright permission on Copyright Marketplace
KEYWORDS
Photodynamic therapy

Skin

Dermatology

Light

Light sources

Tissues

Tumors

Back to Top