review photodynamic therapy with the phthalocyanine … · 2008-09-08 · photodynamic therapy with...

10
Review Photodynamic therapy with the phthalocyanine photosensitizer Pc 4: The case experience with preclinical mechanistic and early clinicaltranslational studies Janine D. Miller a,d,e , Elma D. Baron a,d,e,f , Heather Scull a,d,e , Andrew Hsia a , Jeffrey C. Berlin a,c , Thomas McCormick a,d , Valdir Colussi b , Malcolm E. Kenney a,e , Kevin D. Cooper a,d,e,f , Nancy L. Oleinick b,d,e, a Department of Dermatology, Case Western Reserve University/University Hospitals of Cleveland, Cleveland, OH 44106, USA b Department of Radiation Oncology, School of Medicine, Case Western Reserve University/University Hospitals of Cleveland, Cleveland, OH 44106, USA c Department of Chemistry, Case Western Reserve University/University Hospitals of Cleveland, Cleveland, OH 44106, USA d The Case Skin Diseases Research Center, Case Western Reserve University/University Hospitals of Cleveland, Cleveland, OH 44106, USA e The Case Comprehensive Cancer Center, Case Western Reserve University/University Hospitals of Cleveland, Cleveland, OH 44106, USA f Louis-Stokes VA Medical Center, 10701 East Boulevard, Cleveland, OH 44106, USA Received 4 October 2006; revised 6 January 2007; accepted 10 January 2007 Available online 15 February 2007 Abstract Photodynamic therapy (PDT) is emerging as a promising non-invasive treatment for cancers. PDT involves either local or systemic administration of a photosensitizing drug, which preferentially localizes within the tumor, followed by illumination of the involved organ with light, usually from a laser source. Here, we provide a selective overview of our experience with PDT at Case Western Reserve University, specifically with the silicon phthalocyanine photosensitizer Pc 4. We first review our in vitro studies evaluating the mechanism of cell killing by Pc 4-PDT. Then we briefly describe our clinical experience in a Phase I trial of Pc 4-PDT and our preliminary translational studies evaluating the mechanisms behind tumor responses. Preclinical work identified (a) cardiolipin and the anti-apoptotic proteins Bcl-2 and Bcl-xL as targets of Pc 4- PDT, (b) the intrinsic pathway of apoptosis, with the key participation of caspase-3, as a central response of many human cancer cells to Pc 4-PDT, (c) signaling pathways that could modify apoptosis, and (d) a formulation by which Pc 4 could be applied topically to human skin and penetrate at least through the basal layer of the epidermis. Clinicaltranslational studies enabled us to develop an immunohistochemical assay for caspase-3 activation, using biopsies from patients treated with topical Pc 4 in a Phase I PDT trial for cutaneous T-cell lymphoma. Results suggest that this assay may be used as an early biomarker of clinical response. © 2007 Elsevier Inc. All rights reserved. Keywords: Photodynamic therapy; Silicon phthalocyanine; Pc 4; Cutaneous T-cell lymphoma; Apoptosis Contents Background: photodynamic therapy .................................................... 291 Overview of the case experience with PDT ................................................ 291 From DNA damage to caspases ...................................................... 292 The role of caspases in PDT-induced apoptosis .............................................. 293 Bcl-2 family proteins in PDT-induced apoptosis ............................................. 293 Signaling pathways and apoptosis in response to Pc 4-PDT ....................................... 294 Available online at www.sciencedirect.com Toxicology and Applied Pharmacology 224 (2007) 290 299 www.elsevier.com/locate/ytaap Corresponding author. Department of Radiation Oncology, School of Medicine, Case Western Reserve University, 10900 Euclid Avenue, Cleveland, OH 44106- 4942, USA. Fax: +1 216 368 1142. E-mail address: [email protected] (N.L. Oleinick). 0041-008X/$ - see front matter © 2007 Elsevier Inc. All rights reserved. doi:10.1016/j.taap.2007.01.025

Upload: others

Post on 03-Aug-2020

6 views

Category:

Documents


0 download

TRANSCRIPT

Page 1: Review Photodynamic therapy with the phthalocyanine … · 2008-09-08 · Photodynamic therapy with the phthalocyanine photosensitizer Pc 4: ... d The Case Skin Diseases Research

Available online at www.sciencedirect.com

Toxicology and Applied Pharmacology 224 (2007) 290–299www.elsevier.com/locate/ytaap

Review

Photodynamic therapy with the phthalocyanine photosensitizer Pc 4:The case experience with preclinical mechanistic and

early clinical–translational studies

Janine D. Miller a,d,e, Elma D. Baron a,d,e,f, Heather Scull a,d,e, Andrew Hsia a,Jeffrey C. Berlin a,c, Thomas McCormick a,d, Valdir Colussi b,

Malcolm E. Kenney a,e, Kevin D. Cooper a,d,e,f, Nancy L. Oleinick b,d,e,⁎

a Department of Dermatology, Case Western Reserve University/University Hospitals of Cleveland, Cleveland, OH 44106, USAb Department of Radiation Oncology, School of Medicine, Case Western Reserve University/University Hospitals of Cleveland, Cleveland, OH 44106, USA

c Department of Chemistry, Case Western Reserve University/University Hospitals of Cleveland, Cleveland, OH 44106, USAd The Case Skin Diseases Research Center, Case Western Reserve University/University Hospitals of Cleveland, Cleveland, OH 44106, USAe The Case Comprehensive Cancer Center, Case Western Reserve University/University Hospitals of Cleveland, Cleveland, OH 44106, USA

f Louis-Stokes VA Medical Center, 10701 East Boulevard, Cleveland, OH 44106, USA

Received 4 October 2006; revised 6 January 2007; accepted 10 January 2007Available online 15 February 2007

Abstract

Photodynamic therapy (PDT) is emerging as a promising non-invasive treatment for cancers. PDT involves either local or systemicadministration of a photosensitizing drug, which preferentially localizes within the tumor, followed by illumination of the involved organ withlight, usually from a laser source. Here, we provide a selective overview of our experience with PDT at Case Western Reserve University,specifically with the silicon phthalocyanine photosensitizer Pc 4. We first review our in vitro studies evaluating the mechanism of cell killing by Pc4-PDT. Then we briefly describe our clinical experience in a Phase I trial of Pc 4-PDT and our preliminary translational studies evaluating themechanisms behind tumor responses. Preclinical work identified (a) cardiolipin and the anti-apoptotic proteins Bcl-2 and Bcl-xL as targets of Pc 4-PDT, (b) the intrinsic pathway of apoptosis, with the key participation of caspase-3, as a central response of many human cancer cells to Pc 4-PDT,(c) signaling pathways that could modify apoptosis, and (d) a formulation by which Pc 4 could be applied topically to human skin and penetrate atleast through the basal layer of the epidermis. Clinical–translational studies enabled us to develop an immunohistochemical assay for caspase-3activation, using biopsies from patients treated with topical Pc 4 in a Phase I PDT trial for cutaneous T-cell lymphoma. Results suggest that thisassay may be used as an early biomarker of clinical response.© 2007 Elsevier Inc. All rights reserved.

Keywords: Photodynamic therapy; Silicon phthalocyanine; Pc 4; Cutaneous T-cell lymphoma; Apoptosis

Contents

Background: photodynamic therapy . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 291Overview of the case experience with PDT . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 291From DNA damage to caspases . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 292The role of caspases in PDT-induced apoptosis . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 293Bcl-2 family proteins in PDT-induced apoptosis . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 293Signaling pathways and apoptosis in response to Pc 4-PDT . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 294

⁎ Corresponding author. Department of Radiation Oncology, School of Medicine, Case Western Reserve University, 10900 Euclid Avenue, Cleveland, OH 44106-4942, USA. Fax: +1 216 368 1142.

E-mail address: [email protected] (N.L. Oleinick).

0041-008X/$ - see front matter © 2007 Elsevier Inc. All rights reserved.doi:10.1016/j.taap.2007.01.025

Page 2: Review Photodynamic therapy with the phthalocyanine … · 2008-09-08 · Photodynamic therapy with the phthalocyanine photosensitizer Pc 4: ... d The Case Skin Diseases Research

291J.D. Miller et al. / Toxicology and Applied Pharmacology 224 (2007) 290–299

The possible role of cardiolipin in cell killing by Pc 4-PDT . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 295Clinical development of Pc 4 in photodynamic therapy at Case Western Reserve University and our translational mechanistic studies. 295

Intravenous Pc 4-PDT trial . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 296Topical Pc 4-PDT trial . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 296

Discussion and conclusions . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 297Acknowledgments . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 298References . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 298

Background: photodynamic therapy

Photodynamic therapy (PDT) is emerging as a promisingnon-invasive treatment for cancers (Babilas et al., 2005;Dougherty et al., 1998). It has been shown to induce favorableresponses in the treatment of cutaneous squamous cell and basalcell carcinomas, as well as cancers of the head, neck, lung,esophagus, and bladder (Wolfsen, 2005; Sibata et al., 2001).This treatment involves either local or systemic administrationof a photosensitizing drug, which preferentially localizes withinthe tumor, followed by illumination of the involved organ withlight, usually from a laser light source. The light excites thephotosensitizing drug, resulting in formation of reactive oxygenspecies, believed to be responsible for the cascade of cellularand molecular events in which the end result is selective tumordestruction (Dougherty et al., 1998).

PDT has many advantages over current cancer treatmentmodalities, such as surgery, chemotherapy, and radiation therapy(Dougherty et al., 1998; Sibata et al., 2001). The treatment isrelatively non-invasive in that it usually requires mere illumina-tion of the tumor site. It does not result in systemicimmunosuppressive effects that would translate to a clinicalopportunistic infection. PDT can also be repeated withoutdetrimental consequences to the patient. However, PDTwith thecurrently FDA-approved photosensitizers is not without adverseeffects. For example, Photofrin®, the first systemic drug to beapproved, is well known for causing an intense inflammatoryand necrotic reaction at the treated site and prolonged wide-spread photosensitivity for up to several weeks post-PDT,thereby imposing severe limitations on the patient's lifestyle(Dougherty et al., 1990; Moriwaki et al., 2001). Because of thisand other drawbacks of Photofrin®, many additional photo-sensitizers have been synthesized, and a few of them havedeveloped into FDA-approved drugs or are in clinical trials.

Notwithstanding the many positive results with PDT, bothpreclinical and clinical protocols are still being optimized toaddress the major reasons that PDT sometimes fails to eradicatethe targeted tumor. Failure generally results from (a) inhomo-geneous delivery of the photosensitizer within the tumor orminimal differential in photosensitizer level between tumor andsurrounding normal tissue, (b) poor penetration of light to someparts of the tumor, and/or (c) inability to ensure that the entiretumor remains sufficiently well oxygenated during the fullphotoirradiation period. To improve light delivery, combina-tions of surface and interstitial illumination have been applied,or PDT has been preceded by tumor-debulking surgery (Vogi etal., 2004). To maintain oxygenation in the face of photo-

chemical depletion of molecular oxygen to generate singletoxygen, the rate of light delivery (the fluence rate) can bereduced, thereby slowing the photodynamic utilization ofoxygen to bring it in line with the diffusion of oxygen fromthe tumor blood supply (Foster et al., 1991; Henderson et al.,2004).

Photosensitizers are usually delivered by intravenous injec-tion and are initially taken up by all tissues; earlier release fromsome normal tissues permits the tumor-to-normal tissue ratio toincrease, but the ratios are rarely sufficient to offer optimalselectivity in the photodynamic effect, and some damage tonormal tissues is inevitable. Furthermore, the retention of somephotosensitizers, especially Photofrin, in the skin after systemicdelivery is responsible for the lingering cutaneous photosensi-tivity experienced by patients undergoing PDT (Vogi et al.,2004; Castano et al., 2006). Topical delivery of a photosensi-tizer to accessible tumors offers the potential for improvementsin selectivity and the elimination of cutaneous photosensitivityat sites distant from the targeted tumor.

Currently, topical PDT using amino-levulinic acid (ALA), orits esters (e.g., methyl-ALA), is used for certain cutaneousmalignancies (e.g., superficial basal cell carcinoma), inflam-matory diseases (e.g., acne), and photorejuvenation (Morton etal., 2002). Advantages of ALA-based PDT are the ease ofadministration of the ALA preparation and selectivity in themetabolism of ALA to the photosensitizer Protoporphyrin IX(PpIX) in malignant cells, pilosebaceous units, and other rapidlygrowing tissue. However, there are at least two disadvantageswith the use of ALA. One disadvantage is the prolonged contactperiod (about 4 to 24 h) necessary before light activation can beperformed. This is because ALA itself is not a photosensitizer,but a precursor that undergoes a biochemical conversion toPpIX. Secondly, significant burning and pain are oftenassociated with ALA-PDT. The pain severity is such thatpatients who have multiple basal cell carcinomas (such as thoseseen in basal cell nevus syndrome) may need to receive thetreatment under general anesthesia.

Now that multiple approvals of several photosensitizers havebeen obtained, and PDT is finding its place in the armamentar-ium for malignant and benign diseases, the next phase ofdevelopment of PDT will derive from understanding themechanisms of how cells and tissues respond after treatment.

Overview of the case experience with PDT

Research on PDT at Case Western Reserve University beganin the mid-1980s with collaborations among faculty in several

Page 3: Review Photodynamic therapy with the phthalocyanine … · 2008-09-08 · Photodynamic therapy with the phthalocyanine photosensitizer Pc 4: ... d The Case Skin Diseases Research

292 J.D. Miller et al. / Toxicology and Applied Pharmacology 224 (2007) 290–299

departments, most notably Chemistry, Dermatology, andRadiation Oncology. These early studies led to the synthesisand characterization of novel silicon phthalocyanines thatproved to be strong, active photosensitizers in cell culturesand model tumors. Phthalocyanines are structurally related toporphyrins but with a larger macrocycle ring system, and hencethey absorb longer (tissue-penetrating) wavelengths of light(Fig. 1). Of the phthalocyanines synthesized and characterized,Pc 4 was selected for intensive study and further development(Oleinick and Evans, 1998).

One of the first contributions of our group to uncovering themechanisms of PDT was the demonstration that PDT was astrong inducer of apoptosis (Agarwal et al., 1991). Interestingly,at that time, the scientific community did not have much interestin or appreciation for the significance of apoptosis in tissuedevelopment or tumor therapy. However, this changed rapidlyover the next few years following the discovery and recognitionof one of the most important proteins controlling apoptosis, Bcl-2 (Raffeld et al., 1987; Tsujimoto et al., 1985; Vaux et al., 1988).Bcl-2 was soon revealed as the founding member of a family ofpro- and anti-apoptotic proteins that are critical regulators of theprimary pathways of apoptosis (Adams and Cory, 1998; Greenand Reed, 1998). By the mid-1990s, it was apparent that thelater steps in apoptosis that were responsible for generatingmorphological and biochemical hallmarks of apoptotic cellswere activated by a series of proteases, now termed caspases(Thornberry and Lazebnik, 1998). Both the Bcl-2 familyproteins and certain caspases proved to be important in definingPDT-induced apoptosis (Oleinick et al., 2002).

The following brief excursion through our experience instudying how PDT kills cells will focus on (a) how we learnedabout the roles of Bcl-2, its homologues, and caspases inapoptosis induced by Pc 4-PDT, (b) our current model for theinitiation of apoptosis by Pc 4-sensitized PDT, and (c) how weare now beginning to apply this knowledge to the evaluation ofhuman cancer treated with Pc 4-PDT.

From DNA damage to caspases

Our earliest exploration into the mechanism of PDT withphthalocyanine photosensitizers focused on DNA damage, inparticular, the formation of DNA single-strand breaks andDNA–protein crosslinks. In one study, we exposed L929 mouse

Fig. 1. The structure of (a) the porphyrin ring

fibroblasts to an LD90 dose of ionizing radiation (i.e., a dosecausing 90% reduction of clonogenic survival) and/or a sub-lethal dose of PDT. Radiation-killed cells remained attached tothe culture dish and repaired the initially produced strand breaksin their DNA. In contrast, cells exposed to both agents becamedetached from the culture dish and showed increased DNAbreakage 1 h after exposure. Therefore, we learned that a doseof PDT that was too low to kill cells on its own was sufficient tocause radiation-killed cells to follow a pathway of morpholo-gical and biochemical changes similar to those killed by PDTalone (i.e., the cells lifted off of the monolayer, and the DNAbecame increasingly fragmented). Although we had no otherevidence at the time, we suggested that this mechanism mightbe apoptosis (Ramakrishnan et al., 1990).

Apoptosis is a physiological mode of cell death that isimportant in normal tissue development and remodeling (Fadeeland Orrenius, 2005). It can also be triggered by many differenttypes of cell stresses, including cancer therapies. Apoptosis ischaracterized by a series of morphological and biochemicalchanges. Of the two major pathways for induction of apoptosis,those triggered internally (intrinsic) and externally (extrinsic),the former is most commonly observed following PDT of cellsin culture (Fig. 2). Early steps in the intrinsic apoptosis pathwayinclude the release of cytochrome c from mitochondria and theloss of mitochondrial membrane potential. This is followed byactivation of a group of intracellular proteases called caspases,which are the primary effector molecules of apoptosis. Caspase-3 is one of the key mediators of apoptosis, and the most widelystudied; therefore, active caspase-3 levels have been used as anindicator of apoptosis (Xue et al., 2001a, 2003a; Whitacre et al.,2002; Wu et al., 2006; Granville et al., 1997). Cleavage ofproteins by caspases ultimately leads to chromatin condensationand the degradation of DNA into oligonucleosome-sizedfragments. All of these steps have been documented in cellsexposed to PDT in vitro (Oleinick et al., 2002).

To directly test for apoptosis, we sized the DNA of PDT-treated murine lymphoma L5178Y cells by agarose gelelectrophoresis and found oligonucleosome-sized DNA frag-ments very early after PDT (Ramakrishnan et al., 1990). In asubsequent study, it was established that caspase activity wasnecessary for the DNA to be cleaved (Xue et al., 2001a; He etal., 1998). It is now clear that the major caspase activated in thecells we have studied is caspase-3. The activation process (a

(b) the phthalocyanine ring, and (c) Pc 4.

Page 4: Review Photodynamic therapy with the phthalocyanine … · 2008-09-08 · Photodynamic therapy with the phthalocyanine photosensitizer Pc 4: ... d The Case Skin Diseases Research

Fig. 2. Main mechanism of induction of apoptosis by Pc 4-PDT in vitro. Primary photodynamic damage to mitochondria causes opening of the permeability transitionpore complex, releasing cytochrome c into the cytosol. There, cytochrome c combines with other cytoplasmic proteins to form the apoptosome, resulting in theactivation of caspases-9 and -3, cleavage of various substrates, such as the inhibitor (ICAD) of the caspase-activated DNase (CAD), fragmentation of DNA, andcondensation of chromatin to generate morphologically apoptotic cells.

293J.D. Miller et al. / Toxicology and Applied Pharmacology 224 (2007) 290–299

specific cleavage of the pro-enzyme form by another caspase,caspase-9), occurs in the apoptosome, a cytosolic complexformed from procaspase-9, procaspase-3, apoptosis-activatingfactor-1 (APAF-1), dATP, and cytochrome c released fromdamaged mitochondria (Fig. 2). Active caspase-3 cleavesnumerous substrates, including the inhibitor of caspase-activated DNase (ICAD), releasing CAD to attack chromatinbetween nucleosomes. Another substrate of caspase-3 is theDNA repair enzyme, poly(ADP-ribose) polymerase (PARP).The cleavage of PARP is also a useful indicator of apoptosis(He et al., 1998). The presence of DNA fragments in cellsundergoing apoptosis is the basis of another commonly usedassay for apoptosis, the TUNEL assay. The TUNEL assay hasbeen used to demonstrate apoptosis in model murine tumorstreated with Pc 4-PDT (Agarwal et al., 1996; Zaidi et al.,1993).

Very early in our studies, we became interested in identifyingthe critical step(s) that trigger apoptosis and/or commit the cellto die following Pc 4-PDT. One way to frame the question wasto ask (a) what role does apoptosis play in the response of cellsto PDT; and (b) what are the earliest biochemical reactionsfollowing the initial photodynamic process?

The role of caspases in PDT-induced apoptosis

Addressing part a, we tested the relevance of the caspase-3-dependent steps in the killing of human breast cancer MCF-7cells (Xue et al., 2001a; Whitacre et al., 2002). MCF-7 cellshave a deletion in the CASP-3 gene and do not expressprocaspase-3 at all. We obtained two derivative cell lines fromDr. C. Froehlich (Northwestern University) that were stablytransfected with human procaspase-3 (MCF-7c3 cells) or theempty vector (MCF-7v cells). These lines were exposed tovarious doses of Pc 4 and light, and several known steps in theapoptosis pathway were measured.

We found that the early steps in apoptosis (loss ofmitochondrial membrane potential and the release of cytochromec from mitochondria into the cytosol) occurred identically in thetwo cell lines. In contrast, activation of caspases-9 and -3,

cleavage of PARP, DNA fragmentation, and formation ofmorphologically apoptotic cells with condensed chromatinwere observed only in the MCF-7c3 cells. As measured bytetrazolium dye reduction (a measure of the number and activityof mitochondria in the cell culture), MCF-7c3 cells were moresensitive to PDT than MCF-7v cells. However, when overall celldeath was measured by a clonogenic assay, the two cell linesdisplayed the same PDT dose dependence. This study revealedthat the activity of caspase-3 was important for the cells to carryout the late steps in apoptosis, but overall cell death was notdependent upon these steps (Xue et al., 2001a).

Bcl-2 family proteins in PDT-induced apoptosis

Addressing part b, we explored the role of Bcl-2 and itshomologues in PDT-induced apoptosis to evaluate the earliestidentifiable biochemical changes post-PDT. Bcl-2 is a 26-kDaprotein that resides in the outer mitochondrial membrane andthe endoplasmic reticulum (ER), anchored via a C-terminaltransmembrane (TM) domain. Bcl-2 contains four BH (Bcl-2homology) domains (BH1-BH4). The other members of thefamily share one or more of these domains. We (Xue et al.,2001b) and the Kessel laboratory (Kim et al., 1999) found thatPDTwith a variety of photosensitizers resulted in the immediateloss of Bcl-2, as observed by western blot analysis of cellularprotein. We subsequently demonstrated that Bcl-xL, a closehomologue of Bcl-2, was also subject to “photodamage”, andthis response involved a tight interaction of these proteins withadjacent membrane proteins and/or lipids, to form largecomplexes that migrate more slowly across the gel duringelectrophoresis relative to the native proteins (Xue et al., 2003a;Usuda et al., 2003a).

Although the precise chemistry of the Bcl-2/xL photoreac-tion is still unclear, several properties suggest its importance inthe cellular response to PDT with Pc 4 and the otherphotosensitizers. First, Bcl-2 photodamage is an immediatephotochemical response to photoactivation of Pc 4, occurringduring photoirradiation, in cells kept on ice, and in the presenceof inhibitors of a wide variety of proteases (Xue et al., 2001b).

Page 5: Review Photodynamic therapy with the phthalocyanine … · 2008-09-08 · Photodynamic therapy with the phthalocyanine photosensitizer Pc 4: ... d The Case Skin Diseases Research

294 J.D. Miller et al. / Toxicology and Applied Pharmacology 224 (2007) 290–299

Thus, the loss of Bcl-2 is not a result of downregulation ordegradation by proteasomes or other cellular proteases. Second,the PDT dose dependence for photodamage closely follows thatfor cell killing, as defined by loss of clonogenicity (Xue et al.,2001b). Third, susceptibility of Bcl-2 to photodamage isdependent upon its transmembrane domain binding to mito-chondria or ER and the presence of at least one of a pair ofalpha-helices between the BH1 and BH2 domains of the protein(Usuda et al., 2003b). These structures (alpha-5 and alpha-6) arethought to form a secondary anchorage site to the membrane.Fourth, overexpression of Bcl-2 can protect cells from cellkilling by PDT; in those cases, higher PDT doses eventuallyphotodamage sufficient amounts of Bcl-2 to overcome theprotection and lead to loss of clonogenicity (Usuda et al.,2003a).

As important as the photodamage to Bcl-2 and Bcl-xLappears to be, it is unlikely to be the sole determining factor inphotocytotoxicity. Evidence has also accumulated for a role ofcertain pro-apoptotic members of the Bcl-2 family. Of these,Bax, Bak, and Bid have received the most attention in PDTstudies (Oleinick et al., 2002). Bax is a cytosolic protein thattranslocates to mitochondria during apoptosis. There, it under-goes a conformational change and oligomerizes, resulting in theopening of the mitochondrial permeability transition porecomplex to allow the release of cytochrome c and other pro-apoptotic factors. Bak, a close homologue of Bax, resides in themitochondrial outer membrane but otherwise appears to carryout the same step in apoptosis as does Bax. Bid is a cytosolicprotein that is cleaved during apoptosis to generate a truncatedprotein, tBid, which then translocates to mitochondria andpromotes the activation of Bax and Bak.

Kessel and co-workers transfected Bcl-2 into human breastepithelial MCF-10A cells and found that cells in which Bcl-2protein was overexpressed also contained increased levels ofBax. When these cells were exposed to PDT, Bcl-2 levels weredecreased through photodamage, resulting in a greatly elevatedratio of Bax to Bcl-2 and greater cell killing than in theuntransfected cells (Kim et al., 1999). Upregulation of Bax uponoverexpression of Bcl-2 does not occur in any of the humancancer cell lines we have studied (Xue et al., 2003b); however,some of these lines (the human prostate cancer DU145 andhuman colon cancer HCT-116 lines) are naturally deficient inBax (Chiu et al., 2003, 2005). We have addressed the role ofBax in PDT-induced apoptosis using the above Bax-negativecells, comparing them to corresponding Bax-positive cells(Chiu et al., 2003, 2005). In both cases, the critical lethal lesionappeared to occur upstream of the step in apoptosis controlledby Bax.

However, because all of these cell lines express Bak, itremained possible that Bak was responsible for susceptibility tophotocytotoxicity when Bax was absent. To address the role ofBak, we have recently turned to murine embryonic fibroblasts,obtaining four cell lines expressing both Bax and Bak, Bax only,Bak only, or neither protein. PDT-induced apoptosis was foundin the two lines expressing Bax but in neither line that wasmissing Bax, indicating that Bax is necessary for apoptosis afterPDTand cannot be replaced by Bak (unpublished observations).

However, all of the cell lines were similarly photosensitivewhen cell killing was evaluated by tetrazolium dye reduction(embryonic fibroblasts cannot be evaluated by clonogenicassay).

Our observations indicate that cells die after Pc 4-PDTwhether or not they express Bax and Bak, whether or not theyexpress caspase-3, and whether or not they are competent forapoptosis. Thus, the commitment to cell death following Pc 4-PDT occurs at an early step in the process. However, in cellswith all of the components needed for apoptosis, themorphological and biochemical changes of apoptosis are veryconvenient for evaluating the response to PDT (Fig. 2).

Another anti-apoptotic Bcl-2 homologue, Mcl-1, reveals adifferent response to Pc 4-PDT. Although it has significanthomology to Bcl-2 and Bcl-xL in the domains important forphotodamage, Mcl-1 is not photodamaged. However, we foundthat it is proteolytically degraded in lymphoid cells undergoingapoptosis but not in human A431 keratinocytes (Xue et al.,2005). The degradation of Mcl-1 occurs in Jurkat and U937cells and is dependent on caspase activity, as revealed by itsinhibition by a broad-spectrum caspase inhibitor. The responseof Mcl-1 is thus an indicator of lymphoid cells undergoingapoptosis. We are currently investigating whether Mcl-1degradation can serve as an indicator of the response ofmalignant and/or normal T-cells to PDT after treatment ofcutaneous T-cell lymphoma.

Signaling pathways and apoptosis in response to Pc 4-PDT

PDT has been found to upregulate numerous signalingpathways (Oleinick and Evans, 1998; Moor, 2000). Forexample, stress kinases such as SAPK/JNK and p38/HOGare strongly activated by PDT and may help to promote apop-tosis (Xue et al., 1999). When Hasan Mukhtar was studying Pc4-PDT at Case, he made several notable contributions to ourunderstanding, including demonstrating the presence ofapoptosis within PDT-treated murine tumors (Agarwal et al.,1996; Whitacre et al., 2000; Colussi et al., 1999; Zaidi et al.,1993). One focus of the research on Pc 4-PDT in the Mukhtarlaboratory at Case was on the potential roles of cell-surfacedeath receptors and cell cycle checkpoints. In A431 epider-moid carcinoma cells, they observed increased expression ofFas within the first hour after Pc 4-PDT followed by emer-gence of Fas in the culture medium (Ahmad et al., 2000). Theyalso found multimerization of Fas within the cells, an increasein FasL and FADD expression, the interaction of FasL withFADD, and the activation of caspase 8. Exposure of the cells toeither a blocking antibody or the general caspase inhibitorzVAD-fmk reduced the level of cell killing detected by theMTT assay within 6 or 12 h post-PDT. These data all sug-gested that activation of the extrinsic FAS-mediated pathwayof apoptosis could augment cell death through the intrinsicpathway.

During the progression of A431 cells into apoptosis, anincrease in cells in the G0-G1 phase at the expense of cells in Sphase was noted (Ahmad et al., 1998). These changes werepreceded by an increase in p21/WAF1/CIP1 and downregulation

Page 6: Review Photodynamic therapy with the phthalocyanine … · 2008-09-08 · Photodynamic therapy with the phthalocyanine photosensitizer Pc 4: ... d The Case Skin Diseases Research

295J.D. Miller et al. / Toxicology and Applied Pharmacology 224 (2007) 290–299

of cyclins D1 and E and cyclin-dependent kinases cdk2 and 6.Thus, it appeared that Pc 4-PDT-treated A431 cells experienceda delay in passing the G1/S cell cycle checkpoint. Exploringsubsequent steps in regulating that checkpoint, Ahmad et al.found a decrease in hyperphosphorylated pRb and down-regulation of five members of the E2F family of transcriptionfactors and their heterodimeric partners DP1 and DP2 (Ahmad etal., 1999). The timing of the responses was consistent with adelay at the G1 checkpoint prior to entering the final stages ofapoptosis.

The possible role of cardiolipin in cell killing by Pc 4-PDT

Returning to our earlier question concerning the immediatecritical reaction following PDT that commits cells to apoptosis(or cell death by another mechanism), we now describe somerecent observations implicating mitochondrial lipids as keymediators of the initial photodynamic damage. In an attemptto use the high-affinity ligand of cardiolipin, nonyl-acridineorange (NAO), as a probe of cardiolipin oxidation, we weresurprised to observe that NAO could undergo fluorescenceresonance energy transfer (FRET) with Pc 4 (Morris et al.,2003). The observation of FRET from NAO to Pc 4 indicatesthat these two molecules must reside in mitochondria veryclose to one another (calculated as 7 nm). Because NAO bindsspecifically to cardiolipin, if Pc 4 is close to NAO, it mustalso be very close to cardiolipin. This phospholipid containshighly unsaturated fatty acids and is found only in themitochondrial inner membrane and at the contact sitesbetween the inner and outer membranes. Therefore, cardioli-pin may be one of the immediate targets of singlet oxygen

Fig. 3. Mitochondrial targets of Pc 4-PDT. Pc 4 may bind to both the outer and innewithin proximity of cardiolipin and key components of the mitochondrial inner memopening of the permeability transition pore complex formed by the voltage-depenPhotodamage to Bcl-2 may be an independent event or secondary to lipid oxidation

generated from photoactivation of Pc 4. In fact, Kriska et al.(2005) have demonstrated that cardiolipin, as well as othermembrane phospholipids, can be oxidized by PDT sensitizedby protoporphyrin IX.

What makes cardiolipin such an intriguing target is that itplays an important role in the anchoring of the respiratorycomplexes and cytochrome c to the mitochondrial innermembrane. When cardiolipin is oxidized, cytochrome c isreleased into the intermembrane space. It was recently reportedthat cytochrome c can catalyze the oxidation of cardiolipin toset itself free of the inner membrane (Kagan et al., 2005). Thus,we hypothesize that a (the?) critical lethal event caused by Pc 4-PDT is the oxidation of the cytochrome c–cardiolipincomplexes on the inner mitochondrial membrane. We arecurrently testing this model (Fig. 3).

Clinical development of Pc 4 in photodynamic therapy atCase Western Reserve University and our translationalmechanistic studies

While the mechanistic details of cell killing by Pc 4-PDTwere being elucidated, steps were taken to bring Pc 4 intoclinical studies. With the help of the National CancerInstitute's (NCI) Drug Decision Network, pre-clinical pharma-cokinetic, toxicity, and efficacy studies were carried out.Following these studies, the NCI made available formulatedGMP (Good Manufacturing Practices)-grade Pc 4 for use inphase I clinical trials. Subsequently an Investigational NewDrug Application was approved and a phase I trial opened atthe Case Comprehensive Cancer Center for treatment ofcutaneous cancers.

r mitochondrial membranes. Colocalization of Pc 4 and NAO probably occursbrane. Oxidation of cardiolipin may result in release of cytochrome c throughdent anion channel (VDAC) and the adenine-nucleotide translocator (ANT)..

Page 7: Review Photodynamic therapy with the phthalocyanine … · 2008-09-08 · Photodynamic therapy with the phthalocyanine photosensitizer Pc 4: ... d The Case Skin Diseases Research

296 J.D. Miller et al. / Toxicology and Applied Pharmacology 224 (2007) 290–299

Intravenous Pc 4-PDT trial

This first phase I trial of Pc 4-PDT involved intravenousadministration of the photosensitizer followed 24 h later by laserirradiation of the involved skin sites. Preliminary findingsrevealed that the drug was well tolerated and early in the doseescalation of the trial, one patient with cutaneous T-celllymphoma (CTCL) had a partial response to Pc 4-PDT.However, because accrual to this trial proved difficult, weexplored the possibility of delivering Pc 4 topically to humanskin lesions. Accordingly, we tested a variety of vehicles (e.g.,olive oil, acetone, DMSO, ethanol, propylene glycol) for topicalapplication of Pc 4 and found a combination that was capable ofdelivering the photosensitizer through the stratum corneum. Ourdata using human skin keratome biopsies showed thatformulation of Pc 4 at concentrations ranging from 0.01 mg/mL to 0.1 mg/mL in an ethanol/propylene glycol vehiclepermitted penetration of Pc 4 at least through the basal layer ofthe epidermis within 1 h of application (Swick et al., 2004).

Topical Pc 4-PDT trial

With that information in hand, a phase I clinical trial oftopically applied Pc 4 to treat cutaneous malignancies wasapproved by the FDA, the Institutional Review Board ofUniversity Hospitals of Cleveland, and the Case Comprehen-sive Cancer Center. This trial is still accruing patients, andresults will be reported elsewhere once it is complete.However, some patients have consented to provide biopsiesfor additional translational mechanistic studies. The availabilityof these biopsies has allowed us to investigate a variety ofassays of PDT response, including those for apoptosis, in orderto choose one or more that can be applied reliably to humanskin biopsies in subsequent trials as a surrogate marker ofactivity. We hope to test the hypothesis that selective apoptosisof the target malignant cell population correlates with clinicalimprovement.

Because our pre-clinical results had indicated that T-cellsappear to be more susceptible to Pc 4-PDT-induced apoptosisthan were keratinocytes (Swick et al., 2004), we reasoned thatcutaneous T-cell lymphoma (CTCL), mycosis fungoides (MF)type, might be an appropriate malignancy to target. Ourreasoning centered around epidermotropism, an elevatedconcentration of malignant T-cells in the epidermis, which isa cardinal diagnostic feature of MF-type CTCL. The MF type isthe most common form of CTCL and accounts for almost 50%of all primary cutaneous lymphomas (Kim et al., 1999;Willemze et al., 2005; Willemze and Meijer, 2006). Our centerhas an active multidisciplinary cutaneous lymphoma program,providing access to a well-characterized cohort of patients.Thus, patients with early-stage MF were included in thetranslational methodological study. These patients wereenrolled in the topical Pc 4-PDT phase I clinical trial currentlyunderway at University Hospitals of Cleveland. Early-stagedisease was considered to include TNM classification IA, IB,and IIA. The patients were concurrently not using any othertype of systemic or topical medication for their disease.

Patients were evaluated clinically pre-treatment, and threeareas of involved skin were chosen for treatment (maximum5 cm×5 cm each area). These lesions were measured clinicallyby a graded 5-point scale, via chromometer, via ultrasound,and by global evaluation. Additional lesions were alsoevaluated in the same manner and served as control untreatedlesions. Treated lesions were selected for biopsy along withuntreated lesions that were matched both anatomically andmorphologically.

The Pc 4 (NSC 676418) drug supply utilized in this studywas provided by NCI/CTEP. An amount of the formulated Pc4 sufficient to cover the skin area to be treated (approximately10 μL/cm2) was pipetted onto the lesion surface, and appliedby an investigator via a finger cot. Because this is a phase Istudy, the Pc 4 dose applied (0.01, 0.05, or 0.10 mg/mL) andlight fluence were escalated sequentially in groups of threepatients each. If no toxicity was noted at the lowest drug andlight doses, the light dose was escalated in the next threepatients until 100 J/cm2 was reached, and then the drug dosewas escalated while returning to the lowest light dose again.This will be repeated until a maximally toxic dose (MTD) isreached.

The treated areas were covered with plastic wrap and anopaque dressing for 1 h. After removal of the dressing, visiblelight of 675 nm was delivered (50–100 J/cm2; 100 mW/cm2)from an Applied Optronics diode laser (AOC MedicalSystems, South Plainfield, NJ) coupled to a fiber optic cableterminating in a microlens. The entire Pc 4-treated area wasirradiated. Twenty-four hours after treatment, both the treatedand selected untreated (control) lesions of patients wereevaluated using the same criteria, and two 6-mm biopsieswere taken, one from a treated lesion, and one from an adjacentuntreated lesion. The specimens were fixed in formalin forroutine H&E and immunohistochemical staining. Based on ourprevious experience in detecting apoptotic T-cells in psoriaticskin induced by broad-band, narrow-band, or monochromaticUVB-radiation, we focused on TUNEL and caspase staining.TUNEL staining resulted in excessive background signal,whereas anti-active caspase-3 immunohistochemistry showspromise in this application.

The fixed biopsies were embedded in paraffin, and seriallycut into 4-μm sections. The sections were mounted onto poly-L-lysine-coated slides and deparaffinized. After epitope retrievaland blocking, the sections were incubated with polyclonal anti-active caspase-3 antibody (dilution 1:250; Cayman Chemical,Ann Arbor, MI), followed by biotinylated anti-rabbit immu-noglobulin, and then Vectastain ABC (avidin–biotin–perox-idase complex) (Vector Laboratories, Burlingame, CA). Areaspositive for caspase-3 induction stained brown after develop-ment with diaminobenzidine. Slides were counterstained withMayer's Hematoxylin (Sigma-Aldrich, St. Louis, MO), rinsedwith distilled water, allowed to dry, and then mounted forviewing purposes. Five 20× brightfield images were taken fromeach section using an AxioCam HR digital camera andAxioPhot microscope (Carl Zeiss, Germany).

To validate this technique, we preliminarily examinedbiopsies from a patient with a clear clinical response, and

Page 8: Review Photodynamic therapy with the phthalocyanine … · 2008-09-08 · Photodynamic therapy with the phthalocyanine photosensitizer Pc 4: ... d The Case Skin Diseases Research

Fig. 4. Immunohistochemical identification of cells with caspase-3 protein. Biopsy sections are stained blue-purple with hematoxylin and brown for caspase-3. Foursections are shown: (a) untreated non-responder, (b) treated non-responder, (c) untreated responder, and (d) treated responder.

297J.D. Miller et al. / Toxicology and Applied Pharmacology 224 (2007) 290–299

from a patient without a clinical response. All sections re-vealed some brown-stained areas that were considered caspase-3-positive against the blue-purple hematoxylin staining.Sample images from both treated and untreated lesions areshown in Fig. 4. The caspase-3 positivity of the untreatedlesions was similar for the responding and non-respondingpatient. By contrast, for both patients, the treated lesionsshowed greater caspase-3 positivity than did the matcheduntreated lesions. Of interest, however, was that the increase incaspase-3 positivity was markedly greater in the sections fromthe therapy-responsive patient's lesion (Fig. 4). The images areconsistent with an increase in apoptosis in responding lesions,and serve to validate the use of anti-active caspase-3 for furthertesting in the clinical trial as a biomarker of PDT activity invivo, and hopefully, as an early surrogate marker of clinicalresponse.

In regard to clinical response, our preliminary experienceis promising, although the trial is only partially complete. Ofthe 30 patients treated thus far in the phase I trial of topicalPc 4-PDT, no local or systemic toxicities have been reported.Even though only a single treatment was delivered to eachlesion in this study, a subset of patients responded to therapyshowing improvement in the Pc 4-PDT-treated lesions, asevidenced by thinning and decreased erythema. Both theresponders and non-responders tolerated the treatment well,with no side effects or pain reported. We noted that theresponders consistently reported a minor tingling sensationduring laser irradiation.

Discussion and conclusions

Topical application is a convenient and safe method ofspecifically directing photosensitizers to accessible lesions,avoiding the widespread distribution that occurs followingintravenous administration. This concept has been well studiedfor the administration of the porphyrin prodrugs, ALA and itsesters. However, until now, little attention has been given tothe delivery of active photosensitizers after the apparent failureof studies with porphyrins (Brown et al., 2004). Our clinicalstudy suggests that topically applied Pc 4 can be efficacious forCTCL and perhaps other dermal malignancies, such as basalcell carcinoma, as well as non-malignant conditions of theskin, such as psoriasis. In the appropriate vehicle, Pc 4 mayalso be efficiently delivered into certain other accessiblecancers, such as those of the head-and-neck, esophagus, orlung.

As mentioned in Background: photodynamic therapy, PDThas certain advantages over surgery for the removal of skincancers, including focusing the damage on the tumor cells ratherthan on the collagen matrix; the presence of undisturbed matrixpromotes re-epithelialization of the area with less scarring thanproduced by surgery (Triesscheijn et al., 2006). In addition tobeing much less invasive than surgery, PDT with a topicallyapplied photosensitizer in our protocol is less intrusive in apatient's life, less painful, and has a shorter recovery time. Animportant advantage of PDT over other cancer treatments is theability to attack tumors through direct damage to the malignant

Page 9: Review Photodynamic therapy with the phthalocyanine … · 2008-09-08 · Photodynamic therapy with the phthalocyanine photosensitizer Pc 4: ... d The Case Skin Diseases Research

298 J.D. Miller et al. / Toxicology and Applied Pharmacology 224 (2007) 290–299

cells, damage to the tumor vasculature that secondarily killsmalignant cells, and promotion of inflammatory and immuneresponses that can eliminate malignant cells missed by the firsttwo mechanisms (Castano et al., 2006). In fact, PDT is beingstudied as a means to generate anti-tumor immunity by avaccine-type mechanism (Gollnick et al., 2002; Korbelik, 2006).The ease of administration and potential efficacy of PDT withtopically applied Pc 4 makes this modality attractive fortreatment of skinmalignancies and for evaluating the importanceof all three types of mechanism.

Apoptosis is commonly observed in cells in vitro as well as inmany animal tumor models exposed to PDT with numerousphotosensitizers. Although caspase-3 activity is not essential forcell death after PDT, it is activated in most human cancer cellsand is a very useful measure of the induction of apoptosis. Thereis little information and very few studies on the incidence orrelevance of apoptosis to human tumors responding to PDT(Oleinick et al., 2002; Lilge et al., 2000). Although PDT hasbeen found to be beneficial in treating cancers and other skinconditions (Oleinick et al., 2002; Lui et al., 2004; Bissonnette etal., 2002), there is a lack of studies evaluating the mechanismbehind improvement in skin lesions following PDT. It is also notknown how well the incidence of apoptosis correlates with theresponse to PDT of patients with skin cancer. The preliminaryresults of our phase I clinical trial have been positive, andsuggest that apoptosis may correlate with clinical improvementfollowing Pc 4-PDT.

Acknowledgments

Research on PDT in our laboratories has been supported bythe following NIH grants: R01 CA83917 (to NLO), R01 CA-106491 (to NLO), P01 CA48735 (to NLO), P30 CA43703, AR-39750 (to KDC), T32 AR-007569 (to KDC), R01 AR-051498(to KDC).

References

Adams, J.M., Cory, S., 1998. The bcl-2 protein family: arbiters of cell survival.Science 281, 1322–1326.

Agarwal, M.L., Clay, M.E., Harvey, E.J., Evans, H.H., Antunez, A.R., Oleinick,N.L., 1991. Photodynamic therapy induces rapid cell death by apoptosis inL5178Y mouse lymphoma cells. Cancer Res. 51, 5993–5996.

Agarwal, R., Korman, N.J.,Mohan, R.R., et al., 1996. Apoptosis is an early eventduring phthalocyanine photodynamic therapy-induced ablation of chemi-cally induced squamous papillomas in mouse skin. Photochem. Photobiol.63, 547–552.

Ahmad, N., Feyes, D.K., Agarwal, R., Mukhtar, H., 1998. Photodynamictherapy results in induction of WAF1/CIP1/p21, leading to cell cycle arrestand apoptosis. Proc. Natl. Acad. Sci. U. S. A. 95, 6977–6982.

Ahmad, N., Gupta, S., Mukhtar, H., 1999. Involvement of retinoblastoma (Rb)and E2F transcription factors during photodynamic therapy of humanepidermoid carcinoma cells A431. Oncogene 19, 1891–1896.

Ahmad, N., Gupta, S., Feyes, D.K., Mukhtar, H., 2000. Involvement of fas(APO-1/CD-95) during photodynamic-therapy-mediated apoptosis in humanepidermoid carcinoma cells. J. Invest. Dermatol. 115, 1041–1046.

Babilas, P., Karrer, S., Sidoroff, A., Landthaler, M., Szeimies, R.M., 2005.Photodynamic therapy in dermatology—An update. Photodermatol. Photo-immunol. Photomed. 21, 142–149.

Bissonnette, R., Tremblay, J.F., Juzenas, P., Boushira, M., Lui, H., 2002.

Systemic photodynamic therapy with aminolevulinic acid induces apoptosisin lesional T lymphocytes of psoriatic plaques. J. Invest. Dermatol. 119,77–83.

Brown, S.B., Brown, E.A., Walker, I., 2004. The present and future role ofphotodynamic therapy in cancer treatment. Lancet Oncol. 5, 497–508.

Castano, A.P., Mroz, P., Hamblin, M.R., 2006. Photodynamic therapy and anti-tumour immunity. Nat. Rev. Cancer 6, 535–545.

Chiu, S.M., Xue, L.Y., Usuda, J., Azizuddin, K., Oleinick, N.L., 2003. Bax isessential for mitochondrion-mediated apoptosis but not for cell death causedby photodynamic therapy. Br. J. Cancer 89, 1590–1597.

Chiu, S.M., Xue, L.Y., Azizuddin, K., Oleinick, N.L., 2005. Photodynamictherapy-induced death of HCT 116 cells: apoptosis with or without baxexpression. Apoptosis 10, 1357–1368.

Colussi, V.C., Feyes, D.K., Mulvihill, J.W., et al., 1999. Phthalocyanine 4 (Pc 4)photodynamic therapy of human OVCAR-3 tumor xenografts. Photochem.Photobiol. 69, 236–241.

Dougherty, T.J., Cooper, M.T., Mang, T.S., et al., 1990. Cutaneous phototoxicoccurrences in patients receiving photofrin. Lasers Surg. Med. 10, 485–488.

Dougherty, T.J., Gomer, C.J., Henderson, B.W., et al., 1998. Photodynamictherapy. J. Natl. Cancer Inst. 90, 889–905.

Fadeel, B., Orrenius, S., 2005. Apoptosis: a basic biological phenomenon withwide-ranging implications in human disease. J. Intern. Med. 258, 479–517.

Foster, T.H., Murant, R.S., Bryant, R.G., Knox, R.S., Gibson, S.L., Hilf, R.,1991. Oxygen consumption and diffusion effects in photodynamic therapy.Radiat. Res. 126, 296–303.

Gollnick, S.O., Vaughan, L., Henderson, B.W., 2002. Generation of effectiveantitumor vaccines using photodynamic therapy. Cancer Res. 62,1604–1608.

Granville, D.J., Levy, J.G., Hunt, D.W., 1997. Photodynamic therapy inducescaspase-3 activation in HL-60 cells. Cell Death Differ. 4, 623–628.

Green, D.R., Reed, J.C., 1998. Mitochondria and apoptosis. Science 281,1309–1312.

He, J., Whitacre, C.M., Xue, L.Y., Berger, N.A., Oleinick, N.L., 1998. Proteaseactivation and cleavage of poly(ADP-ribose) polymerase: an integral part ofapoptosis in response to photodynamic treatment. Cancer Res. 58, 940–946.

Henderson, B.W., Gollnick, S.O., Snyder, J.W., et al., 2004. Choice ofoxygen-conserving treatment regimen determines the inflammatory res-ponse and outcome of photodynamic therapy of tumors. Cancer Res. 64,2120–2126.

Kagan, V.E., Tyurin, V.A., Jiang, J., et al., 2005. Cytochrome c acts as acardiolipin oxygenase required for release of proapoptotic factors. Nat.Chem. Biol. 1, 223–232.

Kim, H.R., Luo, Y., Li, G., Kessel, D., 1999. Enhanced apoptotic response tophotodynamic therapy after bcl-2 transfection. Cancer Res. 59, 3429–3432.

Korbelik, M., 2006. PDT-associated host response and its role in the therapyoutcome. Lasers Surg. Med. 38, 500–508.

Kriska, T., Korytowski, W., Girotti, A.W., 2005. Role of mitochondrialcardiolipin peroxidation in apoptotic photokilling of 5-aminolevulinate-treated tumor cells. Arch. Biochem. Biophys. 433, 435–446.

Lilge, L., Portnoy, M., Wilson, B.C., 2000. Apoptosis induced in vivo byphotodynamic therapy in normal brain and intracranial tumour tissue. Br. J.Cancer 83, 1110–1117.

Lui, H., Hobbs, L., Tope, W.D., et al., 2004. Photodynamic therapy of multiplenonmelanoma skin cancers with verteporfin and red light-emitting diodes:two-year results evaluating tumor response and cosmetic outcomes. Arch.Dermatol. 140, 26–32.

Moor, A.C., 2000. Signaling pathways in cell death and survival afterphotodynamic therapy. J. Photochem. Photobiol., B 57, 1–13.

Moriwaki, S.I., Misawa, J., Yoshinari, Y., Yamada, I., Takigawa, M., Tokura, Y.,2001. Analysis of photosensitivity in Japanese cancer-bearing patientsreceiving photodynamic therapy with porfimer sodium (photofrin). Photo-dermatol. Photoimmunol. Photomed. 17, 241–243.

Morris, R.L., Azizuddin, K., Lam, M., et al., 2003. Fluorescence resonanceenergy transfer reveals a binding site of a photosensitizer for photodynamictherapy. Cancer Res. 63, 5194–5197.

Morton, C.A., Brown, S.B., Collins, S., et al., 2002. Guidelines for topicalphotodynamic therapy: report of a workshop of the British photodermato-logy group. Br. J. Dermatol. 146, 552–567.

Page 10: Review Photodynamic therapy with the phthalocyanine … · 2008-09-08 · Photodynamic therapy with the phthalocyanine photosensitizer Pc 4: ... d The Case Skin Diseases Research

299J.D. Miller et al. / Toxicology and Applied Pharmacology 224 (2007) 290–299

Oleinick, N.L., Evans, H.H., 1998. The photobiology of photodynamic therapy:cellular targets and mechanisms. Radiat. Res. 150, S146–S156.

Oleinick, N.L., Morris, R.L., Belichenko, I., 2002. The role of apoptosis inresponse to photodynamic therapy: what, where, why, and how. Photochem.Photobiol. Sci. 1, 1–21.

Raffeld, M., Wright, J.J., Lipford, E., et al., 1987. Clonal evolution of t(14;18)follicular lymphomas demonstrated by immunoglobulin genes and the18q21 major breakpoint region. Cancer Res. 47, 2537–2542.

Ramakrishnan, N., Clay, M.E., Friedman, L.R., Antunez, A.R., Oleinick, N.L.,1990. Post-treatment interactions of photodynamic and radiation-inducedcytotoxic lesions. Photochem. Photobiol. 52, 555–559.

Sibata, C.H., Colussi, V.C., Oleinick, N.L., Kinsella, T.J., 2001. Photodynamictherapy in oncology. Expert Opin. Pharmacother. 2, 917–927.

Swick, A., Camouse, M., McCormick, T., et al., 2004. Successful penetration oftopically-applied silicon phthalocyanine photosensitizer Pc 4 and new Pc 4salts into human skin. J. Invest. Dermatol. 122, A146.

Thornberry, N.A., Lazebnik, Y., 1998. Caspases: enemies within. Science 281,1312–1316.

Triesscheijn, M., Baas, P., Schellens, J.H., Stewart, F.A., 2006. Photodynamictherapy in oncology. Oncologist 11, 1034–1044.

Tsujimoto, Y., Cossman, J., Jaffe, E., Croce, C.M., 1985. Involvement of the bcl-2 gene in human follicular lymphoma. Science 228, 1440–1443.

Usuda, J., Azizuddin, K., Chiu, S.M., Oleinick, N.L., 2003a. Association betweenthe photodynamic loss of bcl-2 and the sensitivity to apoptosis caused byphthalocyanine photodynamic therapy. Photochem. Photobiol. 78, 1–8.

Usuda, J., Chiu, S.M., Murphy, E.S., Lam, M., Nieminen, A.L., Oleinick, N.L.,2003b. Domain-dependent photodamage to bcl-2. A membrane anchorageregion is needed to form the target of phthalocyanine photosensitization.J. Biol. Chem. 278, 2021–2029.

Vaux, D.L., Cory, S., Adams, J.M., 1988. Bcl-2 gene promotes haemopoieticcell survival and cooperates with c-myc to immortalize pre-B cells. Nature335, 440–442.

Vogi, T.J., Eichler, K., Mack, M.G., et al., 2004. Interstitial photodynamic lasertherapy in interventional oncology. EUR Radiol. 14, 1063–1073.

Whitacre, C.M., Feyes, D.K., Satoh, T.H., et al., 2000. Photodynamic therapywith the phthalocyanine photosensitizer Pc 4 of SW480 human colon cancerxenografts in athymic mice. Clin. Cancer Res. 6, 2021–2027.

Whitacre, C.M., Satoh, T.H., Xue, L., Gordon, N.H., Oleinick, N.L., 2002.Photodynamic therapy of human breast cancer xenografts lacking caspase-3.Cancer Lett. 179, 43–49.

Willemze, R., Jaffe, E.S., Burg, G., et al., 2005. WHO-EORTC classification forcutaneous lymphomas. Blood 105, 3768–3785.

Willemze, R., Meijer, C.J., 2006. Classification of cutaneous T-cell lymphoma:from alibert to WHO-EORTC. J. Cutan. Pathol. 33 (Suppl. 1), 18–26.

Wolfsen, H.C., 2005. Uses of photodynamic therapy in premalignant andmalignant lesions of the gastrointestinal tract beyond the esophagus. J. Clin.Gastroenterol. 39, 653–664.

Wu, Y., Xing, D., Luo, S., Tang, Y., Chen, Q., 2006. Detection of caspase-3activation in single cells by fluorescence resonance energy transferduring photodynamic therapy induced apoptosis. Cancer Lett. 235,239–247.

Xue, L.Y., He, J., Oleinick, N.L., 1999. Promotion of photodynamic therapy-induced apoptosis by stress kinases. Cell Death Differ. 6, 855–864.

Xue, L.Y., Chiu, S.M., Oleinick, N.L., 2001a. Photodynamic therapy-induceddeath of MCF-7 human breast cancer cells: a role for caspase-3 in the latesteps of apoptosis but not for the critical lethal event. Exp. Cell Res. 263,145–155.

Xue, L.Y., Chiu, S.M., Oleinick, N.L., 2001b. Photochemical destruction of theBcl-2 oncoprotein during photodynamic therapy with the phthalocyaninephotosensitizer Pc 4. Oncogene 20, 3420–3427.

Xue, L.Y., Chiu, S.M., Oleinick, N.L., 2003a. Staurosporine-induced death ofMCF-7 human breast cancer cells: a distinction between caspase-3-dependent steps of apoptosis and the critical lethal lesions. Exp. Cell Res.283, 135–145.

Xue, L.Y., Chiu, S.M., Fiebig, A., Andrews, D.W., Oleinick, N.L., 2003b.Photodamage to multiple Bcl-xL isoforms by photodynamic therapy withthe phthalocyanine photosensitizer Pc 4. Oncogene 22, 9197–9204.

Xue, L.Y., Chiu, S.M., Oleinick, N.L., 2005. Differential responses of Mcl-1 inphotosensitized epithelial vs. lymphoid-derived human cancer cells.Oncogene 24, 6987–6992.

Zaidi, S.I., Oleinick, N.L., Zaim, M.T., Mukhtar, H., 1993. Apoptosis duringphotodynamic therapy-induced ablation of RIF-1 tumors in C3H mice:electron microscopic, histopathologic and biochemical evidence. Photo-chem. Photobiol. 58, 771–776.