+ All Categories
Home > Documents > Intratumor Administration of the Photosensitizer Pc 4 Affords Photodynamic Therapy Efficacy and...

Intratumor Administration of the Photosensitizer Pc 4 Affords Photodynamic Therapy Efficacy and...

Date post: 24-Jan-2017
Category:
Upload: soumya
View: 212 times
Download: 0 times
Share this document with a friend
7
Intratumor Administration of the Photosensitizer Pc 4 Affords Photodynamic Therapy Efficacy and Selectivity at Short Drug-Light Intervals 1 Thomas H. Foster*, Benjamin R. Giesselman*, Rui Hu , Malcolm E. Kenney and Soumya Mitra* *Department of Imaging Sciences, University of Rochester, Rochester, NY, USA; Department of Biostatistics and Computational Biology, University of Rochester, Rochester, NY, USA; Department of Chemistry, Case Western Reserve University, Cleveland, OH, USA Abstract We evaluated intratumor (IT) versus intravenous (IV) administration of the photosensitizer Pc 4 with respect to tu- mor photosensitizer concentration, specificity, and responses to irradiation. BALB/c mice bearing intradermal EMT6 tumors were given 0.3 mg/kg Pc 4 injected IT or IV through the tail vein. Photosensitizer concentration was evaluated by chloroform extraction and localization assessed by fluorescence imaging and spectroscopy in vivo. Tumors were irradiated at 667 nm, 50 mW/cm 2 , and 100 J/cm 2 . Cures were defined as no palpable tumor 90 days after irradiation. Tumor Pc 4 concentrations 1 hour after IT administration were 35,000-fold higher than measured 24 hours after IV administration (0.112 vs 0.317 × 10 5 μg Pc 4/mg tumor). Exquisite tumor selectivity was observed 1 hour after IT injection. Fluorescence imaging of freshly sectioned tumors revealed no regions de- void of sensitizer at this time point, with pixel intensities in a midline section within a factor of 3 of the peak in- tensity. For identical photosensitizer doses, IT administration significantly improved tumor responses to irradiation, with more than 70% of tumors cured with ITPc 4PDT. In this model, ITPc 4 administration provides improved tumor control, greater selectivity, and opportunity for a short drug-light interval. Translational Oncology (2010) 3, 135141 Introduction Intravenous (IV) and topical routes of photosensitizer administration are widely used in clinical and preclinical applications of photo- dynamic therapy (PDT). Topical application of various formulations of the prodrug aminolevulinic acid (ALA) is routinely performed for superficial lesions in the skin [1]. ALA has also been administered orally for PDT of tumors not accessible to topical delivery [2,3] and for fluorescence-guided surgical tumor resection [4]. Direct intratumor (IT) delivery of photosensitizers has been inves- tigated only sporadically, and the previous literature on this subject is relatively sparse and conflicted. Kostron et al. [5] compared intraperi- toneal (IP) and IT injection of hematoporphyrin derivative (HPD) in subcutaneous and in intracerebral gliosarcomas in a rat model. They reported three- to four-fold increases in tritiated HPD at both tumor sites after IT versus IP administration. Selectivity was also enhanced significantly. The increased HPD concentrations in tumors after di- rect IT injection resulted in improved photodynamic inactivation of the tumor cells, as measured using in vivo and in vitro clonogenic assays. In a pair of articles published in 1988, Amano et al. [6] and Lin et al. [7] also reported results of experiments designed to evaluate IT delivery of HPD. Motivated primarily by the persistent skin photosensitivity that results from systemic administration of HPD and a number of other photosensitizers, these authors dem- onstrated HPD concentrations in a subcutaneous mouse bladder tu- mor line 3 to 15 times higher with IT relative to IP injection [6]. Increased tumor HPD concentrations did not, however, result in im- proved tumor cell killing under the conditions of this study [7], which was attributed to a possible lack of efficient tumor blood vessel photosensitization with IT administration. Gibson et al. [8] investi- gated IT delivery of Photofrin II in a transplanted subcutaneous rat mammary tumor. Photosensitizer levels were not measured directly. Using an in vivo in vitro protocol, in which Photofrin was injected in vivo through an IT or IP route and tumor and liver mitochondria Address all correspondence to: Thomas H. Foster, PhD, Department of Imaging Sciences, University of Rochester Medical Center, 601 Elmwood Ave, Box 648, Rochester, NY 14642. E-mail: [email protected] 1 This work was supported by the National Institutes of Health grant CA122093 awarded by the National Cancer Institute. Received 1 October 2009; Revised 5 November 2009; Accepted 11 November 2009 Copyright © 2010 Neoplasia Press, Inc. All rights reserved 1944-7124/10/$25.00 DOI 10.1593/tlo.09295 www.transonc.com Translational Oncology Volume 3 Number 2 April 2010 pp. 135141 135
Transcript
Page 1: Intratumor Administration of the Photosensitizer Pc 4 Affords Photodynamic Therapy Efficacy and Selectivity at Short Drug-Light Intervals

Intratumor Administrationof the Photosensitizer Pc 4Affords Photodynamic TherapyEfficacy and Selectivity atShort Drug-Light Intervals1

Thomas H. Foster*, Benjamin R. Giesselman*,Rui Hu†, Malcolm E. Kenney‡ and Soumya Mitra*

*Department of Imaging Sciences, University of Rochester,Rochester, NY, USA; †Department of Biostatistics andComputational Biology, University of Rochester, Rochester,NY, USA; ‡Department of Chemistry, Case Western ReserveUniversity, Cleveland, OH, USA

AbstractWe evaluated intratumor (IT) versus intravenous (IV) administration of the photosensitizer Pc 4 with respect to tu-mor photosensitizer concentration, specificity, and responses to irradiation. BALB/c mice bearing intradermalEMT6 tumors were given 0.3 mg/kg Pc 4 injected IT or IV through the tail vein. Photosensitizer concentrationwas evaluated by chloroform extraction and localization assessed by fluorescence imaging and spectroscopyin vivo. Tumors were irradiated at 667 nm, 50 mW/cm2, and 100 J/cm2. Cures were defined as no palpable tumor90 days after irradiation. Tumor Pc 4 concentrations 1 hour after IT administration were 35,000-fold higher thanmeasured 24 hours after IV administration (0.112 vs 0.317 × 10−5 μg Pc 4/mg tumor). Exquisite tumor selectivitywas observed 1 hour after IT injection. Fluorescence imaging of freshly sectioned tumors revealed no regions de-void of sensitizer at this time point, with pixel intensities in a midline section within a factor of 3 of the peak in-tensity. For identical photosensitizer doses, IT administration significantly improved tumor responses to irradiation,with more than 70% of tumors cured with IT–Pc 4–PDT. In this model, IT–Pc 4 administration provides improvedtumor control, greater selectivity, and opportunity for a short drug-light interval.

Translational Oncology (2010) 3, 135–141

IntroductionIntravenous (IV) and topical routes of photosensitizer administrationare widely used in clinical and preclinical applications of photo-dynamic therapy (PDT). Topical application of various formulationsof the prodrug aminolevulinic acid (ALA) is routinely performed forsuperficial lesions in the skin [1]. ALA has also been administeredorally for PDT of tumors not accessible to topical delivery [2,3]and for fluorescence-guided surgical tumor resection [4].Direct intratumor (IT) delivery of photosensitizers has been inves-

tigated only sporadically, and the previous literature on this subject isrelatively sparse and conflicted. Kostron et al. [5] compared intraperi-toneal (IP) and IT injection of hematoporphyrin derivative (HPD) insubcutaneous and in intracerebral gliosarcomas in a rat model. Theyreported three- to four-fold increases in tritiated HPD at both tumorsites after IT versus IP administration. Selectivity was also enhancedsignificantly. The increased HPD concentrations in tumors after di-rect IT injection resulted in improved photodynamic inactivation ofthe tumor cells, as measured using in vivo and in vitro clonogenicassays. In a pair of articles published in 1988, Amano et al. [6]and Lin et al. [7] also reported results of experiments designed toevaluate IT delivery of HPD. Motivated primarily by the persistent

skin photosensitivity that results from systemic administration ofHPD and a number of other photosensitizers, these authors dem-onstrated HPD concentrations in a subcutaneous mouse bladder tu-mor line 3 to 15 times higher with IT relative to IP injection [6].Increased tumor HPD concentrations did not, however, result in im-proved tumor cell killing under the conditions of this study [7],which was attributed to a possible lack of efficient tumor blood vesselphotosensitization with IT administration. Gibson et al. [8] investi-gated IT delivery of Photofrin II in a transplanted subcutaneous ratmammary tumor. Photosensitizer levels were not measured directly.Using an in vivo – in vitro protocol, in which Photofrin was injectedin vivo through an IT or IP route and tumor and liver mitochondria

Address all correspondence to: Thomas H. Foster, PhD, Department of ImagingSciences, University of Rochester Medical Center, 601 Elmwood Ave, Box 648, Rochester,NY 14642. E-mail: [email protected] work was supported by the National Institutes of Health grant CA122093awarded by the National Cancer Institute.Received 1 October 2009; Revised 5 November 2009; Accepted 11 November 2009

Copyright © 2010 Neoplasia Press, Inc. All rights reserved 1944-7124/10/$25.00DOI 10.1593/tlo.09295

www.transonc.com

Trans la t iona l Onco logy Volume 3 Number 2 April 2010 pp. 135–141 135

Page 2: Intratumor Administration of the Photosensitizer Pc 4 Affords Photodynamic Therapy Efficacy and Selectivity at Short Drug-Light Intervals

preparations were subsequently irradiated in vitro, activities of twomitochondrial enzymes were attenuated to a significantly greater de-gree when irradiation of tumor samples was performed 2 hours afterIT injection. Irradiation of tumor and liver mitochondrial preparationsat 2 hours after IT Photofrin injection showed tumor selectivity, withactivities of enzymes in liver mitochondria 5- to 10-fold less susceptibleto PDT-mediated damage. This study also showed increased efficacy ofPhotofrin PDT as measured by tumor growth delay when irradiationwas performed 2 hours after Photofrin injection IT versus IP.IT delivery of the photosensitizer methylene blue has received

slightly more attention in the literature, in part because it is reducedto a colorless, photodynamically inactive form when injected system-ically [9]. König et al. [10] showed significant reduction in tumormass in small subcutaneous Ehrlich carcinomas in mice in responseto methylene blue administered through several IT injections andmultiple laser irradiations delivered in daily fractions. Two articlesby Orth et al. [11,12] described the efficacy of IT–methyleneblue–PDT in colorectal tumors in mice. Their latter article reportedno evidence of tumor in 79% of mice 5 weeks after an initial PDTtreatment, with a second methylene blue injection and irradiationperformed at 2 weeks if the tumor had not been eradicated by thefirst treatment. Encouraging results were reported from a small pilotstudy in three esophageal cancer patients [13]. Methylene blue wasinjected directly into inoperable, recurrent carcinomas under endo-scopic guidance, and laser irradiation was performed 1 hour later.After two treatments within a 2-week period, there were no adverseside effects and no macroscopic evidence of tumor.Very recently, two reports have evaluated ITmeso-tetrahydroxyphenyl

chlorin (mTHPC, Foscan) administration in rodent models of breast[14] and brain [15] cancer. Both studies were motivated by the pro-longed skin photosensitivity associated with this promising photosen-sitizer. D’Hallewin et al. [14] used the liposomal formulation, Foslip,and showed maximum IT mTHPC fluorescence and irradiation-induced tumor necrosis at a 24-hour drug-light interval. This surpris-ingly long optimal interval was attributed to concentration quenchingwithin liposomes and a relatively long rate of redistribution, thusdrawing attention to the importance of the sensitizer formulation.In an intracranial glioma model in rats, Mannino et al. [15] foundcomparable tumor uptake and tumor-to-normal tissue ratios after IPand IT Foscan delivery in an ethanol/ethylene glycol (40:60) vehiclebut found that favorable results were obtained in the IT case with20-fold lower administered photosensitizer dose and a shorter(4 hours) drug-light interval.Despite some promising initial studies, IT photosensitizer adminis-

tration has not been pursued systematically and it remains an under-studied area with significant clinical potential. The purpose of ourstudy was to perform the first investigation of the IT delivery ofthe second-generation photosensitizer Pc 4, a topical formulationof which has been evaluated in a phase 1 clinical trial for the treatmentof cutaneous T-cell lymphoma [16]. Here we describe extremely highsensitizer concentrations, excellent tumor selectivity, and long-termcures using IT–Pc 4–PDTwith a short 1-hour drug-light interval.

Materials and Methods

Animal and Tumor ModelMouse mammary EMT6 tumors were initiated on the backs of

female BALB/c mice by the intradermal (ID) injection of 106 cells.

Animals were followed daily to track tumor growth and were fed ex-clusively on a chlorophyll-free diet prepared according to the recipeof Holmes et al. [17] to eliminate chlorophyll-derived fluorescence.Approximately 7 to 10 days after implantation when the tumorsreached a volume of approximately 25 to 40 mm3, they were usedfor control, PDT treatment, or drug extraction studies. For in vivoimaging of host cell infiltration and perfusion, tumors were initiatedby the injection of 5 × 105 EMT6 cells into the ID space of the earpinna [18].

Photosensitizer Administration and Light TreatmentThe silicon phthalocyanine photosensitizer, Pc 4 [16], was prepared

as described previously [19]. Pc 4 was dissolved in a 50% ethanol–50% Cremophor solution, and a stock concentration of 2.1 mg/mlwas prepared. The stock was diluted in a ratio of 1:9 in 0.9% salineto a final concentration of 0.21 mg/ml. The volume for IT injectionwas approximately 35 μl, which was injected at a single point nearthe center of the tumor using a 29-gauge needle. For IV injection,35 μl of 0.21 mg/ml was further diluted in 65 μl of 90% saline,5% ethanol, and 5% Cremophor. To facilitate direct comparison, thedose of administered Pc 4 through either IT or IV route was main-tained at 0.3 mg/kg. After 1 hour and 24 hours of drug-light inter-vals for IT and IV administration, respectively, the tumors weresubjected to PDT irradiation using 667-nm light from a diode laser(Power Technology, Inc, Little Rock, AR). Light was deliveredthrough a GRIN-lens-terminated multimode fiber (OZ Optics,Ottawa, Ontario, Canada), and the tumors were illuminated witha fluence of 100 J/cm2 at an irradiance of 50 mW/cm2. Controlsincluded untreated ((−) drug, (−) light) animals and mice receivingonly 0.3 mg/kg Pc 4 IT (no light).

Drug ExtractionPc 4 levels in the tumor after IV and IT injection were quantified

using chloroform extraction. To accomplish this, 0.3 mg/kg Pc 4 wasinjected either IT or IV. Either 1 hour (IT) or 24 hours (IV) afterinjection, the mouse was euthanized and the tumor was excised.For mice that received IT injection, the skin immediately adjacentto the tumor was also harvested. Tumors were weighed and subjectedto chloroform extraction (D. Kessel, personal communication). First,tumors were homogenized in a tissue grinder containing 1 ml ofHank’s balanced salt solution per 10 mg of tumor. The amount0.8 ml of MeOH per 10 mg of tumor was added to the solutionand vortexed; next, 0.8 ml of chloroform per 10 mg of tumor wasadded. The solution was then centrifuged at 227g for 10 minutesto allow for the separation of the aqueous and chloroform phases.The bottom layer consisting of the chloroform and the solubilizeddrug was transferred to a cuvette, and absorbance and fluorescencemeasurements were performed for IT and IV administration, respec-tively. The Pc 4 concentration was calculated from these measurementsusing a calibration curve generated from the absorbance and fluores-cence peak amplitudes of known Pc 4 concentrations in the same sol-vent. Skin sections were frozen in liquid nitrogen for 30 minutes andpowdered with a frozen pestle before homogenization. Otherwise, theywere treated identically to tumor sections.

Photosensitizer Fluorescence Imaging and SpectroscopyPhotosensitizer distribution and degree of tumor localization were

assessed through in vivo imaging. One hour after IT–Pc 4 injection,

136 PDT with Intratumor Administration of Pc 4 Foster et al. Translational Oncology Vol. 3, No. 2, 2010

Page 3: Intratumor Administration of the Photosensitizer Pc 4 Affords Photodynamic Therapy Efficacy and Selectivity at Short Drug-Light Intervals

anesthetized mice were positioned on the stage of a stereofluorescencemicroscope (Model SMZ1500; Nikon Instruments, Melville, NY)equipped with an Xcite illumination source (EXFO, Ontario, Canada)and a computer-controlled x-y translation stage (H101A ProScanII;Prior Scientific, Rockland, MA). Excitation of Pc 4 and fluorescencecollection were accomplished using a custom filter cube (HQ560/120x;635DCXR;HQ645LPm,ChromaTechnology, Rockingham,VT). In-dividual fields acquired using a 0.5× objective with a 0.75× magnifica-tion zoom corresponded to fields of view (FOVs) of 25.2mm × 19mm.Images of entire mice were constructed by translating the stage andstitching individual fields to create a montage (MetaVue 6.1; MolecularDevices, Sunnyvale, CA).To examine the detailed spatial distribution of Pc 4 in the tumors

and immediately adjacent normal skin, 1 hour after injection, micewere euthanized and tumors were excised immediately. Tumors weresectioned mediolaterally, and the resulting fresh section was imagedwith the stereofluorescence microscope (0.5× objective, 2× zoom).Skin was left on the excised tumor tissue to provide spatial orientation.High-resolution images of 1390 × 1040 pixels with an FOVof 9.5mm×7 mm were captured and digitized by a Photometrics 12-bit mono-chrome CCD camera (CoolSNAPHQ; Roper Scientific, Inc, Trenton,NJ). To assess sensitizer heterogeneity, image analysis was performed onthe tumor region using the Surface Plot tool in ImageJ (NIH; URL:http://rsb.info.nih.gov/ij/).In vivo fluorescence spectroscopy measurements were made using a

system and custom multiple-optical-fiber probe described previously[20]. Briefly, Pc 4 fluorescence was excited using a 639-nm laser(Power Technology), and broadband reflectance was acquired usinga white light source (75-W xenon arc lamp, model 6263; Oriel In-struments, Stratford, CT). Light from these sources and the fluores-cence and reflectance signals were transmitted to/from the tissuesurface through dedicated fibers in the probe. Signals were imagedonto a TE-cooled, 16-bit, 512 × 512 pixel CCD camera (Pixis512;Princeton Instruments, Princeton, NJ) through an imaging spectro-graph (SpectraPro 275; Acton Research Corp, Acton, MA). A long-passfilter (650AELP; Omega Optical, Brattleboro, VT) in the detectionpath was positioned to reject excitation light. The signal measured bythe CCD was first corrected for instrument response.To minimize the possibly confounding effects of tissue optical

properties on the measured fluorescence, the fluorescence spectrumwas divided by the reflectance measured in the same wavelength in-terval and source-detector geometry. Measurements were obtained atseveral locations on the tumor as well at both legs. Before measure-ment, the tumor site and leg areas were shaved, and hair was removedusing a commercial depilating agent (Nair; Church & Dwight, Co,Princeton, NJ).

Tumor Response Assay and StatisticsAfter PDT, tumor dimensions along three orthogonal axes were

measured daily using digital calipers. Volumes were computed assum-ing an approximately ellipsoidal shape with the expression, V = (4/3)πr1r2r3. Mice were removed from the study if the volume of thetumor reached twice the pretreatment volume. Cures were definedas no evidence of palpable tumor 90 days after PDT. Statistical anal-ysis was performed using pairwise comparisons of tumor regrowthcurves among control, Pc 4–only, IV–PDT, and IT–PDT treatmentgroups. A log-rank test was applied to each pair using SAS 9.1 (SASInstitute, Inc, Cary, NC). Bonferroni adjustments were applied tothe raw P values of the log-rank tests to guard against type I error.

Host Response and Vascular Perfusion Imaging After PDTIn vivo confocal imaging of host responses or perfusion status in

tumors of live mice was performed using a custom laser scanning fluo-rescence confocal microscope as described previously [21]. Briefly,anesthetized mice were placed in a supine position with the ear tumorsfacing the top of a coverslip mounted on the stage of the inverted mi-croscope. The fluorophore Alexa Fluor 647 was excited with a 639-nmdiode laser and detected using a 647-nm long-pass filter (Semrock,Rochester, NY). Alexa Fluor 488 and fluorescein isothiocyanate (FITC)were excited at 488 nm from an argon ion laser and detected usinga combination of 500-nm long-pass and 515/30 nm band-pass filters(Chroma Technology). The combination of a 100-μm-diameter pin-hole and a 10×, 0.45 NA objective gave a 6-μm optical section thick-ness, and the images were acquired at 16 bits with a lateral resolutionof 1 μm per pixel.For immunofluorescence imaging in vivo [18,22], antibodies were

purchased directly as fluorescent conjugates. We labeled neutrophil in-filtration into tumors using ID administration of antimouse GR-1antibodies (clone RB6-8C5; Biolegend, San Diego, CA). The vesselsin the tumor were labeled by ID injection of antimouse CD31 anti-bodies (clone MEC13.3; Biolegend). The ID injection volumes of anti-bodies were approximately 30 μl and concentrations were 0.1 mg/ml.Antibodies were administered as a cocktail 3 hours before imaging toallow for clearance of unbound label. To visualize perfusion status intumors, 200 μl of 5 mg/ml FITC-dextran (FD2000S; Sigma-Aldrich,St Louis, MO) was injected IV through the tail vein. Perfusion inCD31-positive Alexa Fluor 647–labeled tumor vasculature was imagedas early as 5 minutes after injection.

Results and DiscussionPreliminary imaging of Pc 4 fluorescence in vivo indicated that Pc 4was distributed throughout the tumor within less than 1 hour afterIT injection. Published studies of Pc 4–PDT in mice typically used a24-hour drug-light interval with IV administration of 0.6 mg/kg [23].Thus, our comparisons were made on the basis of these two intervals.Anticipating favorable Pc 4 concentrations with IT injection, we choseto compare IT versus IV routes of administration using a two-foldlower concentration of 0.3 mg/kg. Pc 4 levels in freshly excised tumorswere evaluated using chloroform extraction 1 or 24 hours after IT orIV administration, respectively. As shown in Table 1, the mean Pc 4concentration recovered from the tumor 1 hour after IT injection wasapproximately 35,000-fold greater than that extracted 24 hours afterIV injection of the same Pc 4 concentration. Because IT administra-tion resulted in some accumulation in the skin immediately adjacent tothe ID tumor, we measured Pc 4 concentration there as well. Greaterthan 95% of the injected Pc 4 was recovered from tumor and imme-diately adjacent skin, with approximately 65.7% of the injected sensi-tizer extracted from the tumor and 32.0% from the skin.

In vivo, whole-mouse fluorescence imaging showed a high degreeof localization of Pc 4 in tumors and overlying skin within 1 hour of

Table 1.Mean (±SD) Pc 4 Concentration in Tumors at 1 and 24 Hours afterIT and IV Administration, Respectively, as Quantified by ChloroformExtraction.

IT – 1 hour (n = 8) IV – 24 hours (n = 6)

0.112 (±0.025) 0.317 × 10−5 (±0.707 × 10−6)

Concentrations are expressed as micrograms of Pc 4 per milligram of tumor.

Translational Oncology Vol. 3, No. 2, 2010 PDT with Intratumor Administration of Pc 4 Foster et al. 137

Page 4: Intratumor Administration of the Photosensitizer Pc 4 Affords Photodynamic Therapy Efficacy and Selectivity at Short Drug-Light Intervals

IT injection at a single point (Figure 1). Image analysis revealed fluo-rescence counts from the normal skin adjacent to the tumor that wereat or very near control levels at this time point. Optical-fiber–basedpoint fluorescence spectroscopy is more sensitive than imaging to lowfluorophore levels. Thus, we used this technique to evaluate the sys-temic distribution of Pc 4 to a site remote from the tumor, the leg ofthe mouse. At 1 hour after IT injection, singular-value decomposi-tion analysis [20] of spectra like those shown in Figure 2A indicatedapproximately 50-fold greater Pc 4 fluorescence intensity at the tumorrelative to the leg. Spectroscopy measurements performed in mice24 hours after IV administration showed comparable, low fluorescenceemission from the tumor and the remote site, reflecting little if anytumor selectivity (not shown). Pc 4 undergoes modest irradiation-induced photobleaching in human cutaneous T-cell lymphoma le-sions [24]. We observed photobleaching in the ID murine tumorsas well as shown in Figure 2B. Remarkably, spectroscopy at the tumorsite 24 hours after irradiation showed complete recovery of the Pc 4fluorescence (Figure 2B). Because there is no evidence that the photo-bleaching of Pc 4 is reversible, we attribute this increase in fluorescenceto the monomerization of initially aggregated, concentration-quenchedsensitizer, a phenomenon we have observed previously in monolayercell culture [25]. Photobleaching followed by more modest increasesin Pc 4 absorption 4 hours after irradiation of tumors sensitized with2 mg/kg Pc 4 IV was reported recently by Bai et al. [26].A concern with IT sensitizer delivery is the possibility of extremely

heterogeneous IT distribution. To address this concern directly, weevaluated Pc 4 fluorescence distribution 1 hour after IT injectionin freshly excised tumor sections using stereofluorescence micros-copy. As illustrated in the representative image of Figure 3A, no gra-dient relative to the point of injection was observed. An analysis ofimage pixel intensities shown in Figure 3B revealed an approximatelythree-fold differential between the maximum and minimum fluores-cence counts, with fluorescence in all pixels significantly above con-trol levels. Interestingly, although chloroform extraction revealed atwo-fold greater Pc 4 concentration in the tumor relative to the im-mediately adjacent skin, this is not reflected in the fluorescence re-

corded in the freshly excised sections. This again is consistent withsignificant concentration quenching in the tumor, as noted above.Tumor growth control was used to compare the therapeutic effi-

cacy of IT versus IV–Pc 4 administration with an irradiation regimenconsisting of 100 J/cm2 delivered at an irradiance of 50 mW/cm2.The Kaplan-Meier curves of Figure 4 demonstrate that the tumor re-sponse to Pc 4–PDTand ITadministration was dramatically enhancedrelative to that observed with IV injection of the same photosensitizerconcentration. Among the 15 mice treated with IT administration,11 (73%) were cured as defined by no evidence of tumor 90 days afterirradiation. No cures were observed in the IV group (n = 8), where themedian tumor volume doubling time was 6 days. The difference intumor control between IV– and IT–PDTwas highly significant, witha Bonferroni-adjusted P < .001. Untreated controls ((−) light, (−) drug)displayed a median tumor doubling time of 4 days (n = 6), whereasmice that received Pc 4 injections IT but were not irradiated ((−) light,(+) drug) showed a modest but statistically significant growth delayof 10 days (P = .02, n = 5). Tumor regrowth in response to IV–Pc4–PDT was statistically indistinguishable from that in response toIT–Pc 4 injection without irradiation (P = 1.0).

Figure 1. Fluorescence image illustrating Pc 4 localization in an IDtumor 1 hour after IT injection at a single location. The whole mouseimage was created from a series of adjacent stereofluorescenceimages, each with 25.2 mm× 19mm FOVs, which were montagedafter acquisition as described in Materials and Methods. Because ofthe intense, highly localized fluorescence at the tumor site, the dis-play is saturated at the tumor to render the rest of the mouse vis-ible. The inset shows the individual field containing the tumor. Herethe display has been rescaled to eliminate saturation. The brightcentral region corresponds to the tumor.

Figure 2. (A) Representative fluorescence spectra obtained fromtumor and remote leg in vivo 1 hour after IT–Pc 4 administration.(B) Fluorescence spectra from the tumor site before, immediatelyafter, and 24 hours after irradiation (100 J/cm2), as indicated in thelegend. In all cases, spectra were acquired with 639 nm excitation,and the fluorescence amplitudes correspond to an acquisitiontime of 0.1 second.

138 PDT with Intratumor Administration of Pc 4 Foster et al. Translational Oncology Vol. 3, No. 2, 2010

Page 5: Intratumor Administration of the Photosensitizer Pc 4 Affords Photodynamic Therapy Efficacy and Selectivity at Short Drug-Light Intervals

As noted above, Pc 4 was found in the skin immediately adjacentto the ID tumor. PDTresulted in eschar formation directly above thetumor, as shown in Figure 5. The skin healed well with completeregrowth of hair within approximately 3 weeks after irradiation.When photosensitizers are administered IV, PDT induces a local

inflammatory response that is characterized by leukocyte infiltration,with a significant fraction of these infiltrating cells being neutrophils[27–29]. To examine the extent of this response to IT–Pc 4–PDT, weimaged the influx of GR-1+ neutrophils in tumors in vivo 24 hours

after irradiation. Figure 6, A and B, illustrates the fluorophore-labeledinfiltrating neutrophils imaged in an untreated control and PDT-treated tumor, respectively. Irradiation resulted in a significant three-to five-fold (n = 4) enhanced accumulation of GR-1+ cells at this timepoint. To the best of our knowledge, these results represent the firstdemonstration of a PDT-induced inflammatory response visualized di-rectly in vivo through noninvasive optical imaging, and they representthe first characterization of any kind of an acute inflammatory re-sponse to Pc 4–PDT.Perfused tumor blood vessels are required for the survival and re-

growth of clusters of PDT-treated tumor cells that escape immediatephototoxicity, but they may also have an important function in facil-itating the trafficking of host cells into and out of the treated tumor.This complex scenario serves to illustrate the importance of the perfu-sion status of vessels and its role in influencing mechanisms of long-term tumor response.With this motivation, we imaged tumor perfusionin live mice before and 24 and 48 hours after IT–Pc 4–PDT usingIV-injected FITC-conjugated high–molecular weight dextran as anoptical perfusion marker [30]. As illustrated in the representative imagesof Figure 6, C and D, there was no detectable difference in perfusionstatus between the control and treated tumors, thus suggesting thatIT–Pc 4–PDT does not initiate functional damage to the vascula-ture, at least up to 48 hours after irradiation.

Figure 3. (A) Stereofluorescence image of Pc 4 distribution in afresh, excised tumor section. The tumor was excised 1 hour afterIT injection of 0.3 mg/kg Pc 4. The white circle bounds the ID tu-mor, and the bright regions outside of the circle correspond to theskin adjacent to the tumor, where Pc 4 concentrations were halfthat within the tumor. (B) A surface plot of pixel intensities fromthe tumor region of A. The Pc 4 fluorescence counts within thetumor ranged from approximately 300 to 1000.

Figure 4. Kaplan-Meier curves of tumor responses to IT–Pc 4–PDT.Laser irradiation at 667 nm was performed at an irradiance of50 mW/cm2 for a fluence of 100 J/cm2 1 or 24 hours after IT orIV administration of 0.3 mg/kg Pc 4. Improved efficacy of IT– versusIV–Pc 4 was highly statistically significant (P < .001).

Figure 5. Digital photographs of the treatment site and surrounding tissue before and at various times after IT–Pc 4–PDT.

Translational Oncology Vol. 3, No. 2, 2010 PDT with Intratumor Administration of Pc 4 Foster et al. 139

Page 6: Intratumor Administration of the Photosensitizer Pc 4 Affords Photodynamic Therapy Efficacy and Selectivity at Short Drug-Light Intervals

The recent study published by Bai et al. [26] offers interestingpoints of comparison with our findings. Using a Pc 4 dose of 2 mg/kgadministered IV and assessing tumor and skin sensitizer concentra-tions with a form of absorption spectroscopy in vivo, those authorsdemonstrated a relationship between tumor Pc 4 levels at the timeof irradiation and tumor response. Their two cures (among 16 PDT-treated mice) after irradiation with 150 J/cm2 occurred in animals withthe highest Pc 4 concentrations, and among the 14 mice whose tumorsregrew, a correlation between tumor growth delay and IT–Pc 4 con-centration was demonstrated. Thus, these data suggest that photosen-sitizer concentration can be a limiting factor in tumor response to PDTin vivo. Of course, it is not possible to arbitrarily increase the amountof systemically administered drug, which makes IT delivery very attrac-tive in those situations where it is clinically feasible. Indeed, it is un-likely that the extremely high IT–Pc 4 concentrations realized in ourexperiments could ever be achieved with systemic administration, evenif toxicity and skin photosensitivity were of no concern.In addition to greatly increased sensitizer concentrations in the tar-

get tissue, IT administration of Pc 4 offers exceptional selectivity. Asobserved also by Bai et al. [26], we found no tumor selectivity up to24 hours after IV Pc 4 delivery. Peak selectivity at 48 hours was amodest 2.1-fold, whereas spectroscopy in our study revealed at least50-fold tumor-to-normal skin selectivity in as little as 1 hour after IT

administration. The ability to achieve high selectivity at very shortdrug-light intervals would be attractive to patients and clinicians, en-abling an entire treatment to be completed within a single, reason-ably short visit. Yet another potential advantage of IT administrationis the opportunity it presents for using less drug per patient, therebylowering the cost of PDT substantially. Finally, taken together, re-duced persistent skin photosensitivity, good IT distribution, andlower cost would facilitate repeat PDT treatments, which would bemore difficult for those photosensitizers that are retained in the skin.Beyond those tumor sites that are obviously accessible to IT injec-tion, the capabilities of modern interventional radiology render manysolid tumors throughout the body candidates for this approach.

AcknowledgmentsThe authors thank David Kessel for detailed advice on the sensitizerextraction protocol.

DisclosureMalcolm E. Kenney is a founder and member of the scientific ad-

visory board of Fluence, a new start-up company seeking to commer-cialize Pc 4 for PDT.

Figure 6. (A, B) In vivo confocal images of Alexa Fluor 488–conjugated anti–GR-1+ neutrophils (green) and Alexa Fluor 647–conjugatedanti-CD31 vessels (red) in EMT6 tumors grown in the ears of a BALB/c mouse. (A) Control untreated tumor and (B) IT–Pc 4–PDT–treatedtumor 24 hours after irradiation. GR-1+ neutrophils and CD31+ vessels were imaged by ID injection of approximately 30 μl of 0.1 mg/mlfluorophore-conjugated antibodies into the ear 3 hours before imaging. (C, D) In vivo confocal fluorescence images of perfusion (green)in CD31-positive vessels (red) in (C) control untreated EMT6 ear tumors and (D) IT–Pc 4–PDT–treated tumor 48 hours after irradiation.Perfusion was imaged by injecting 200 μl of 5 mg/ml FITC–dextran IV through the tail vein, and the perfusion status in CD31+ Alexa Fluor647–labeled tumor vasculature was imaged as early as 5 minutes after injection. The FOV in the images is 800 μm × 800 μm. Imageswere acquired at a depth of approximately 100 μm in the tumor.

140 PDT with Intratumor Administration of Pc 4 Foster et al. Translational Oncology Vol. 3, No. 2, 2010

Page 7: Intratumor Administration of the Photosensitizer Pc 4 Affords Photodynamic Therapy Efficacy and Selectivity at Short Drug-Light Intervals

References[1] Morton CA, McKenna KE, and Rhodes LE (2008).British Association of Derma-

tologists Therapy Guidelines and Audit Subcommittee Guidelines for topicalphotodynamic therapy: update. Br J Dermatol 159, 1245–1266.

[2] Mackenzie GD, Dunn JM, Selvasekar CR, Mosse CA, Thorpe SM, NovelliMR, Bown SG, and Lovat LB (2009). Optimal conditions for successful abla-tion of high-grade dysplasia in Barrett’s oesophagus using aminolaevulinic acidphotodynamic therapy. Lasers Med Sci 24, 729–734.

[3] Beck TJ, Kreth FW, Beyer W, Mehrkens JH, Obermeier A, Stepp H, StummerW, and Baumgartner R (2007). Interstitial photodynamic therapy of nonresect-able malignant glioma recurrences using 5-aminolevulinic acid induced proto-porphyrin IX. Lasers Surg Med 39, 386–393.

[4] Pichlmeier U, Bink A, Schackert G, Stummer W, and the ALA Glioma StudyGroup (2008). Resection and survival in glioblastoma multiforme: an RTOG re-cursive partitioning analysis of ALA study patients. Neuro Oncol 10, 1025–1034.

[5] Kostron H, Bellnier DA, Lin CW, Swartz MR, and Martuza RL (1986). Distribu-tion, retention, and phototoxicity of hematoporphyrin derivative in a rat glioma.Intraneoplastic versus intraperitoneal injection. J Neurosurg 64, 768–774.

[6] Amano T, Prout GR, and Lin C-W (1988). Intratumor injection as a more ef-fective means of porphyrin administration for photodynamic therapy. J Urol139, 392–395.

[7] Lin C-W, Amano T, Rutledge AR, Shulok JR, and Prout GR (1988). Photo-dynamic effect in an experimental bladder tumor treated with intratumor injec-tion of hematoporphyrin derivative. Cancer Res 48, 6115–6120.

[8] Gibson SL, Van Der Meid KR, Murant RS, and Hilf R (1990). Increased efficacyof photodynamic therapy of R3230ACmammary adenocarcinoma by intratumoralinjection of Photofrin II. Br J Cancer 61, 553–557.

[9] DiSanto AR and Wagner JG (1972). Pharmacokinetics of highly ionized drugs.III. Methylene blue–blood levels in the dog and tissue levels in the rat followingintravenous administration. J Pharm Sci 61, 1090–1094.

[10] König K, Bockhorn V, Dietel W, and Schubert H (1987). Photochemotherapyof animal tumors with the photosensitizer methylene blue using a krypton laser.J Cancer Res Clin Oncol 113, 301–303.

[11] Orth K, Russ D, Beck G, Rück A, and Beger HG (1998). Photochemotherapyof experimental colonic tumours with intra-tumorally applied methylene blue.Langenbecks Arch Surg 383, 276–281.

[12] Orth K, Beck G, Genze F, and Rück A (2000). Methylene blue mediated photo-dynamic therapy in experimental colorectal tumors in mice. J Photochem Photo-biol B 57, 186–192.

[13] Orth K, Rück A, Stanescu A, and Berger HG (1995). Intraluminal treatment ofinoperable oesophageal tumours by intralesional photodynamic therapy withmethylene blue. Lancet 345, 519–520.

[14] D’Hallewin MA, Kochetkov D, Viry-Babel Y, Leroux A, Werkmeister E, DumasD, Gräfe S, Zorin V, Guillemin F, and Bezdetnaya L (2008). Photodynamic ther-apy with intratumoral administration of lipid-based mTHPC in a model of breastcancer recurrence. Lasers Surg Med 40, 543–549.

[15] Mannino S, Molinari A, Sabatino G, Ciafrè SA, Colone M, Maira G, Anile C,Arancia G, and Mangiola A (2008). Intratumor vs. systemic administration ofmeta-tetrahydroxyphenylchlorin for photodynamic therapy of malignant glio-

mas: assessment of uptake and spatial distribution in C6 rat glioma model. Int JImmunopathol Pharmacol 21, 227–231.

[16] Miller JD, Baron ED, Scull H, Hsia A, Berlin JC, McCormick T, Colussi V,Kenney ME, Cooper KD, and Oleinick NL (2007). Photodynamic therapywith the phthalocyanine photosensitizer Pc 4: the case experience with preclin-ical mechanistic and early clinical-translational studies. Toxicol Appl Pharmacol224, 290–299.

[17] Holmes HJ, Kennedy JC, Pottier R, Rossi R, and Weagle G (1995). A recipe forthe preparation of a rodent food that eliminates chlorophyll-based tissue fluo-rescence. J Photochem Photobiol B 29, 199.

[18] Cummings RJ, Mitra S, Lord EM, and Foster TH (2008). Antibody-labeled fluo-rescence imaging of dendritic cell populations in vivo. J Biomed Opt 13, 044041.

[19] Anula HM, Berlin JC, Wu H, Li Y-S, Peng X, Kenney ME, and Rodgers MAJ(2006). Synthesis and photophysical properties of silicon phthalocyanines withaxial siloxy ligands bearing alkylamine termini. J Phys Chem 110, 5215–5223.

[20] Finlay JC, Conover DL, Hull EL, and Foster TH (2001). Porphyrin bleachingand PDT-induced spectral changes are irradiance dependent in ALA-sensitizednormal rat skin in vivo. Photochem Photobiol 73, 54–63.

[21] Bigelow CE, Conover DL, and Foster TH (2003). Confocal fluorescence spec-troscopy and anisotropy imaging system. Opt Lett 28, 695–697.

[22] Mitra S and Foster TH (2008). In vivo confocal fluorescence imaging of the intra-tumor distribution of the photosensitizer mono-L-aspartylchlorin-e6. Neoplasia10, 429–438.

[23] Anderson CY, Freye K, Tubesing KA, Li Y-S, Kenney ME, Mukhtar H, andElmets CA (1998). A comparative analysis of silicon phthalocyanine photosen-sitizers for in vivo photodynamic therapy of RIF-1 tumors in C3H mice. Photo-chem Photobiol 67, 332–336.

[24] Lee TK, Baron ED, and Foster TH (2008). Monitoring Pc 4 photodynamictherapy in clinical trials of cutaneous T-cell lymphoma using noninvasive spec-troscopy. J Biomed Opt 13, 030507.

[25] Wang KK-H,Wilson JD, KenneyME,Mitra S, and Foster TH (2007). Irradiation-induced enhancement of Pc 4 fluorescence and changes in light scattering are po-tential dosimeters for Pc 4–PDT. Photochem Photobiol 83, 1–7.

[26] Bai L, Guo J, Bontempo FA, and Eiseman JL (2009). The relationship of Phthalo-cyanine 4 (Pc 4) concentrations measured noninvasively to outcome of Pc 4 photo-dynamic therapy in mice. Photochem Photobiol 85, 1011–1019.

[27] Krosl G, Korbelik M, and Dougherty GJ (1995). Induction of immune cell infil-tration into murine SCCVII tumour by Photofrin-based photodynamic therapy.Br J Cancer 71, 549–555.

[28] Sun J, Cecic I, Parkins CS, and Korbelik M (2002). Neutrophils as inflamma-tory and immune effectors in photodynamic therapy–treated mouse SCCVIItumours. Photochem Photobiol Sci 1, 690–695.

[29] Gollnick SO, Evans SS, Baumann H, Owczarczak B, Maier P, Vaughan L, WangWC, Unger E, and Henderson BW (2003). Role of cytokines in photodynamictherapy–induced local and systemic inflammation. Br J Cancer 88, 1772–1779.

[30] Brown EB, Campbell RB, Tsuzuki Y, Xu L, Carmeliet P, Fukumura D, and JainRK (2001). In vivo measurement of gene expression, angiogenesis and physio-logical function in tumors using multiphoton laser scanning microscopy. NatMed 7, 864–868.

Translational Oncology Vol. 3, No. 2, 2010 PDT with Intratumor Administration of Pc 4 Foster et al. 141


Recommended