You are on page 1of 6

www.elsevier.

nl/locate/jphotobiol

J. Photochem. Photobiol. B: Biol. 52 (1999) 99–104

Tumor blood-flow changes following protoporphyrin IX-based


photodynamic therapy in mice and humans
Mark A. Herman a, David Fromm a,*, David Kessel b
a
Department of Surgery, Wayne State University School of Medicine, 6C-University Health Center, 4201 St. Antoine, Detroit, MI 48201, USA
b
Department of Pharmacology, Wayne State University School of Medicine, 6C-University Health Center, 4201 St. Antoine, Detroit, MI 48201, USA

Received 7 April 1999; accepted 7 September 1999

Abstract

The effects of aminolevulinic acid (ALA)-based photodynamic therapy (PDT) on tumor blood flow are controversial. This study examines
the effects of ALA and Photofrin-based PDT on blood flow of Colon-26 tumors implanted in mice as well as the effects of ALA-based PDT
on blood flow of human colorectal carcinomas and a carcinoid tumor in situ. Tumors are implanted in both flanks of mice. One tumor of each
animal serves as a control. Blood flow is measured using a laser Doppler method. Tumor blood flow in mice not receiving a photosensitizer
but treated with three different light fluences (50, 100 and 150 J/cm2) does not differ significantly from blood flow in the untreated tumor in
the opposite flank. PDT after ALA administration using the three different light fluences does not significantly affect blood flow. In contrast,
PDT after Photofrin administration causes a significant decrease in tumor blood flow with each light fluence, but this change is not as dramatic
as reported in other studies. In contrast to mice, six patients who receive ALA prior to surgery all show a decrease in blood flow (means51.8%,
p-0.001) after PDT using 100 J/cm2. Comparison with other published results suggests that it is likely that flow measurement by the laser
Doppler method underestimates the effects of PDT on tumor blood flow due to the depth of laser penetration. Nevertheless, the present
observations on blood flow suggest that the effects of ALA-based PDT on adenocarcinomas of the colon and rectum as well as an intra-
abdominal carcinoid tumor in humans are more pronounced than would be predicated by some animal studies. q1999 Elsevier Science
S.A. All rights reserved.

Keywords: Photodynamic therapy; Photosensitizers; Tumor blood flow

1. Introduction body [9,11]. However, even less is known about the mech-
anism of cell death following PDT using endogenous pho-
Photodynamic therapy (PDT) of malignant disease tosensitization with ALA. In contrast to Photofrin, the data
involves the administration of a photosensitizing agent and for tumor blood flow after PDT using ALA are controversial
its subsequent activation by light of the appropriate wave- [7,12,13]. Furthermore, it is not known if blood-flow studies
length, resulting in death of the cells containing the agent. of tumors in experimental animals are comparable to those
The precise mechanism of cell death remains uncertain and for human tumors. The purpose of this study was to examine
may be different, depending upon the photosensitizer. the effects of PDT on tumor blood flow of mice after admin-
Among the mechanisms is a decrease in blood flow. Photo- istration of either ALA or Photofrin as well as in human
frin, the most widely used photosensitizer, has been repeatedly patients receiving ALA.
shown to cause tumor vascular shutdown. These studies
implicate ischemia and resultant tumor hypoxia as a primary,
although not necessarily the only, mechanism of cell death 2. Methods
for Photofrin-based PDT [1–8].
5-Aminolevulinic acid (ALA)-induced protoporphyrin IX 2.1. Approvals
(PpIX) is a very attractive photosensitizer from a clinical
standpoint because of its greater tumor specificity compared The animal studies were approved by the Wayne State
with other drugs [9–11] and its rapid clearance from the University School of Medicine Animal Investigation Com-
mittee. The human studies were approved by the Wayne State
* Corresponding author.; e-mail: dfsurg@aol.com University School of Medicine Human Investigation Com-

1011-1344/99/$ - see front matter q1999 Elsevier Science S.A. All rights reserved.
PII S 1 0 1 1 - 1 3 4 4 ( 9 9 ) 0 0 1 0 9 - 8

Thursday Nov 25 01:32 PM StyleTag -- Journal: JPB (J. Photochem. Photobiol. B: Biol.) Article: 7874
100 M.A. Herman et al. / J. Photochem. Photobiol. B: Biol. 52 (1999) 99–104

mittees as well as the FDA. Informed consent was obtained tofrin standards were prepared in a manner identical to the
from all participating patients. specimens.

2.2. Mice 2.3. Humans

2.2.1. Tumors 2.3.1. Tumors


Colon-26 tumor (National Cancer Institute, Frederick Six patients had either isolated or metastatic adenocarci-
Cancer Research and Development Center, DCT Tumor nomas of the colon or rectum and one had a carcinoid tumor.
Repository, Frederick, MD) was subcutaneously implanted Each tumor site used for measurement was at least 2 cm in
in both flanks of 38 female CD2F1 mice (Harlan Sprague thickness. Ages ranged from 34 to 68 years (medians54.5
Dawley, Indianapolis, IN) weighing 20–40 g. The tumors years) and four were women.
were allowed to grow to approximately 0.5 cm=0.5 cm, at
which time the mice were randomly allocated into three
2.3.2. Photosensitizer
groups based on the photosensitizer used.
ALA, 60 mg/kg, (provided by DUSA Pharmaceuticals
Inc., Tarrytown, NY) was mixed in sterile water immediately
2.2.2. Photosensitizers
before use and given as a single bolus orally to six patients
Twelve mice received ALA, 200 mg/kg (Dusa Pharma-
at various intervals prior to PDT. PDT was done during var-
ceuticals, Tarrytown, NY); 14 mice received Photofrin, 10
ious surgical procedures before mobilization of the organ or
mg/kg (QLT Phototherapeutics, Inc., Vancouver, Canada)
tumor to be resected.
and 12 control mice received no drug. All drugs were admin-
istered by tail vein injection. ALA, dissolved in normal saline,
2.3.3. Irradiation
was given 3 h prior to tumor irradiation. Photofrin, dissolved
Each patient received a total tumor fluence of 100 J/cm2
in 5% dextrose, in water, was given 24 h prior to tumor
with an incident fluence rate of 200 mW/cm2 using 633 nm
irradiation. All mice were kept in darkened cages following
light from an argon dye laser (Spectra-Physics, Mountain
photosensitizer administration.
View, CA) delivered through a microlens delivery system
applied to an easily accessible portion of the tumor.
2.2.3. Irradiation
Following anesthesia using sodium pentobarbital, 55 mg/
kg i.p., the tumor in each flank was exposed by elevating a 2.3.4. Tumor photosensitizer detection
flap of overlying skin. One of the two tumors was then Since biopsies to detect tumor concentrations of PpIX in
exposed to 633 nm light from an argon dye laser (Spectra- humans might well alter blood flow, the presence of PpIX
Physics, Mountain View, CA). The opposite tumor, which was detected by a previously reported spectrophotofluoro-
was outside the field of illumination, served as a non-irradi- metric method [15]. The intended site of treatment showed
ated control. Each of the three groups received a total tumor unquestionable accumulation of PpIX in each instance and is
light fluence of 50, 100, or 150 J/cm2. The incident fluence in contrast to surrounding normal tissue.
rate ranged from 55 to 68 mW/cm2. Treatment times thus
varied among groups: 15.2 min to deliver 50 J/cm2, 27.8 min 2.4. Blood-flow measurement
to deliver 100 J/cm2, and 36.6 min to deliver 150 J/cm2.
A laser Doppler flow probe (Laserflo Blood Perfusion
2.2.4. Tumor photosensitizer level Monitor, model BPM 403A, TSI Incorporated, St. Paul, MN)
Concentrations of protoporphyrin IX in colon-26 tumors was used to make quantitative determinations of tumor blood
3 h after the i.v. administration of the same dose of ALA have flow. The probe uses a 780 nm diode laser. This wavelength
been previously reported as 2.3"0.3 mg/gm [14]. is well outside the absorption peaks for both Photofrin and
Photofrin concentrations in tumor were estimated using a PpIX, eliminating drug activation by the flow probe. Blood-
previously described method [11,14]. Tumor samples were flow measurements were made by manually holding the tip
protected from light at all times and stored at y708C prior to of the probe approximately 1 mm from the surface of the
measurement. Briefly, 100 mg samples of tumor were tumor. Since blood flow tended to be heterogeneous, varying
minced, digested over a period of 7–10 days in a 388C incu- from one section of the tumor to another, all serial measure-
bator using a mixture of water and Scintigest (Fisher Scien- ments were taken from the same point on the tumor surface.
tific, Pittsburgh, PA). The samples were next diluted with Baseline tumor blood flow was measured immediately prior
methanol and centrifuged. The fluorescence emission spec- to and at the intended site of light treatment.
trum of the resulting supernatant was measured using broad- Blood flow in mice was measured immediately prior to
band excitation (400"20 nm) with a monochromator and PDT. Following treatment of one tumor in mice, measure-
CCD system (Instaspec IV, Oriel Corp., Stratford, CT). The ments were made from both tumors at 5 min intervals for 30
active porphyrins in Photofrin could easily be distinguished min and subsequently at 45 min, and at 60 min. The animals
from spurious signals arising from other fluorophores. Pho- were then sacrificed and both tumors of the Photofrin animals

Thursday Nov 25 01:32 PM StyleTag -- Journal: JPB (J. Photochem. Photobiol. B: Biol.) Article: 7874
M.A. Herman et al. / J. Photochem. Photobiol. B: Biol. 52 (1999) 99–104 101

were removed for measurement of the concentration of the tumor lying in the opposite flank received no treatment.
photosensitizer. The baseline values for treated and untreated animals are not
Blood-flow measurements in humans were made at a time significantly different within the three groups of mice, Fig.
that vital signs were stable, since induction of anesthesia and 1. However, the mean rates of blood flow between the paired
the infusion of i.v. fluids were kept at a constant rate. Only sets of controls for each light fluence (50, 100 and 150 J/
short intervals of blood flow were measured in humans so as cm2) vary and thus emphasize the importance of measuring
not to interfere with the operative procedure. In each case, blood flow among tumors in each individual animal in order
the tumor was easily exposed so that the blood-flow probe to obtain appropriate control values. The reason(s) for this
could be placed and fixed perpendicular to the tumor. Meas- variability between groups of animals is not clear. Adminis-
urements were made immediately before and immediately tration of a light fluence of 50, 100 or 150 J/cm2 does not
after PDT, which took 8 min. significantly alter blood flow over a 50 min interval, Fig. 1.
2.5. Statistics 3.1.2. ALA
Blood flow prior to treatment with light was compared The baseline blood-flow values for treated and untreated
with the baseline line value obtained from the tumor not mice are not significantly different within the three groups of
treated with light using the paired t-test. The means of all mice. Administration of 50, 100 or 150 J/cm2 to tumors in
subsequent values for treated and untreated animals were also animals treated with ALA does not significantly alter blood
compared using the paired t-test. flow compared with that in the tumor in the opposite flank
that was not exposed to light, Fig. 2. Since the lack of an
3. Results effect on blood flow might be due to limits of detection, the
effect of Photofrin-based PDT was also measured.
3.1. Mice
3.1.3. Photofrin
3.1.1. Controls
The baseline blood-flow values for treated and untreated
One of the two tumors in control animals, who did not
mice are not significantly different within the three groups of
receive a photosensitizer, was treated with the laser whereas

Fig. 1. Effect of three different light fluences (Ns4 animals for each dose) Fig. 2. Effect of three different light fluences (Ns4 animals for each dose)
on blood flow of Colon-26 tumor subcutaneously implanted into both flanks on blood flow of Colon-26 tumor subcutaneously implanted into both flanks
of mice who did not receive a photosensitizer. The tumor in one flank was of mice 3 h after receiving ALA, 200 mg, i.v. The tumor in one flank was
treated with light and the tumor in the opposite flank was not treated with treated with light and the tumor in the opposite flank was not treated with
light. The differences in blood flow are not statistically significant. light. The differences in blood flow are not statistically significant.

Thursday Nov 25 01:32 PM StyleTag -- Journal: JPB (J. Photochem. Photobiol. B: Biol.) Article: 7874
102 M.A. Herman et al. / J. Photochem. Photobiol. B: Biol. 52 (1999) 99–104

Table 1
Tissue concentrations of Photofrin in Colon-26 tumor

Light fluence mg/g tissue


(J/cm2) untreated treated

50 5.6"1.3 5.1"1.1
100 4.6"0.7 3.4"0.7
150 4.0"0.6 3.6"0.4

Light fluences apply only to treated animals. Ns4 in each group.


Valuessmean"SE.

Fig. 3. Effect of three different light fluences (Ns4 animals for 50 J/cm2
and Ns5 animals each for 100 and 150 J/cm2) on blood flow of Colon-26
tumor subcutaneously implanted into both flanks of mice 24 h after receiving
Photofrin, 10 mg/kg, i.v. The tumor in one flank was treated with light and
the tumor in the opposite flank was not treated with light. The differences in
blood flow between tumors treated with light and untreated tumors was
statistically significant for each light dose (see Section 3.1.3).

mice. In contrast to ALA-treated animals, administration of


50, 100 or 150 J/cm2 to tumor in animals treated with Pho- Fig. 4. Effect of 100 J/cm2 light fluence on intra-operative blood flow of
tofrin results in a significant decrease in blood flow at each malignant tissue in situ in six different patients who received ALA, 60 mg/
light fluence (p-0.001 for each light fluence), Fig. 3. How- kg, p.o. Blood flow was measured immediately before and 8 min after PDT.
ever, there does not appear to be a near-complete vascular The origin and location of the tumor as well as the time of PDT after receipt
of ALA is indicated for each patient. The mean decrease in blood flow was
shutdown within 1 h of delivery of any of the three light 51.8"2.7% (p-0.001).
fluences. Tissue concentrations of Photofrin are shown in
Table 1, which indicates accumulation of the drug. changes. These include radiolabeled microsphere injection
[1,2], direct visualization through transparent sandwich
3.2. Humans observation chambers [3], inta-arterial fluorescein dye injec-
Immediately prior to treatment with light, the relative blood tion [4], tumor oxygen tension measurement [5], injection
flow of each tumor was stable, Fig. 4. The blood flow in each and extraction of 86RbCl [6,7], magnetic resonance spec-
patient decreased after treatment with light, Fig. 4. In one troscopy using D2O [16,17], microscopic measurement of
patient, blood flow was measured for 10 min (rectum – changes in blood vessel diameter [12], and ocular angio-
6.5 h, see Fig. 4) and remained depressed at a steady level graphy [18]. Laser Doppler flowmetry is another method
between 30 s and 10 min after treatment. The mean decrease that has been used to measure PDT-related blood-flow
in blood flow (baseline compared with last measurement) changes [4,19–21]. The validity as well as the reliability of
was 51.8"2.7% (p-0.001). this non-invasive method have been confirmed by a number
of studies, but the latter do not involve tissues exposed to
4. Discussion PDT [22–26]. The primary advantage of laser Doppler flow-
metry is that it is an easily performed, real-time quantitation
Previous studies of blood flow following PDT have of tissue blood flow at depths corresponding to laser light
employed differing techniques for measuring vascular penetration.

Thursday Nov 25 01:32 PM StyleTag -- Journal: JPB (J. Photochem. Photobiol. B: Biol.) Article: 7874
M.A. Herman et al. / J. Photochem. Photobiol. B: Biol. 52 (1999) 99–104 103

The 780 nm wavelength of the Doppler will not cause mice has been reported using fluorescein exclusion tests [28].
photoactivation of PpIX or Photofrin, eliminating the possi- In contrast, the present study found no significant effect of
bility of Doppler-induced vascular effects during serial PDT using ALA on the blood flow of colon tumors implanted
recordings. However, there are at least two potential draw- in mice. This lack of response to ALA is not due to the absence
backs of the technique. The first is spatial variation in the of PpIX in the tumor, as we have previously reported signif-
readings. The heterogeneity of tumor tissue combined with icant concentrations of PpIX in such tumors exposed to the
unavoidable hand movements during the manual recordings same dose of ALA and the same time at the start of PDT
can cause changes in blood-flow readings from one area of [14]. Furthermore, it is unlikely that spontaneous changes in
the tumor to another. The data of the present study appear to blood flow (for example, as a result of change in cardiac
suggest that serial recordings made from the same site on output) account for the lack of response, since each animal
each tumor eliminate much of this variability. The second treated with PDT also had blood-flow measurement of the
problem is the depth of tissue penetration by a 780 nm wave- untreated tumor in the opposite flank. While possible, it
length. This is greater than that which occurs with the 633 appears that the lack of response was not due to inadequate
nm laser light source used to activate PpIX and Photofrin. fluence rate or light fluence. Studies finding an effect of ALA-
Thus, Doppler flowmetry carries the possibility of under- based PDT on blood flow used 400 J with 100 mW and 100
estimating changes in tumor blood flow if the laser Doppler J/cm2 with 178 or 300 mW/cm2 [7,12,13], whereas the
light penetrates through areas unaffected by PDT and/or into present study used 100 J/cm2 with 200 mW /cm2 for humans
normal tissues deeper than the tumor. and 55–68 mW/cm2 for mice. Tumor resistance to PDT is
All studies, including the present, that have examined the also unlikely, as other studies have shown that the Colon-26
effects of PDT using Photofrin or hematoporphyrin derivative tumor is responsive to ALA-based PDT [28–30]. It is also
show a decrease in blood flow measured by fluorescein exclu- unlikely that the duration of measurement after PDT was
sion, 86RbCl extraction, microsphere, laser Doppler or micro- inadequate in the present study because virtually all studies
scopic methods. This decrease has been observed for of the effect of PDT on blood flow show an early response.
transplanted fibrosarcoma in mice [6,18] and rats [7], trans- However, the lack of a detectable blood-flow effect of
planted urothelial tumors in rats [1,2], SCCVII cells trans- ALA-based PDT on Colon-26 tumors transplanted in mice
planted in mice [6], isologous mammary tumors implanted and determined by the laser Doppler method must be inter-
in rats [3], and spontaneous mouse mammary tumors as well preted with caution. The majority of animal studies indicate
as transplanted chondrosarcomas in rats [4]. The present that the vascular effects of hematoporphyrin derivatives are
study found a decrease in blood flow in colon tumors dramatic, a situation not observed in the present study. Thus,
implanted in mice. The decrease in blood flow occurring after it is possible that the laser Doppler underestimates flow in
Photofrin-based PDT appears to be independent of the light animal tumors. This may be related to the depth of penetration
fluence. However, another study found that the reduction in of the laser and might account for the lack of a detectable
blood flow is directly proportional to the injected dose of effect of hematoporphyrin derivative on blood flow of human
Photofrin and a more extensive vascular effect with increas- oral cancer transplanted into mice [19].
ing light fluences ranging from 2.4 to 90 J/cm2 [4]. In contrast to the lack of a detectable effect of ALA-based
While most studies to date using exogenously administered PDT in mice, ALA-based PDT in humans with either ade-
photosensitizers have reported a decrease in blood flow, there nocarcinomas of the colon and rectum or a carcinoid tumor
is controversy about the effects of PDT using endogenous is followed by a mean decrease of 52% in blood flow. The
photosensitization following administration of ALA. Exper- duration of this decrease is not clear because of limitations in
imental studies suggest that a vascular effect of ALA-based the time of measurement. However, it is at the very least
PDT does not dominate [12]. Fluorescence microscopic transient, lasting from 21 s to 10 min. This dramatic decrease,
examination also shows low fluorescence in blood vessels of in comparison to the mouse studies, is most likely not due to
dimethylhydrazine-induced colonic tumors in rats 1–8 h after laser Doppler penetration beyond the tumors, because each
i.v. administration of ALA [13]. These data led to the con- measurement was made in tumors that were several centi-
clusion that the primary effect of PDT using ALA was on the meters thick. However, it should be noted that the laser Dopp-
tumor cells rather than the blood vessels. In contrast, another ler measurements in humans may also be an underestimate
fluorescence microscopic study found PpIX in regions imme- of actual changes due to the depth of penetration of the laser.
diately adjacent to tumor microvessels of transplanted rat The only other study involving changes in blood flow in
fibrosarcoma 3 h after administration of ALA, but this study humans due to ALA-based PDT (60 J/cm2 with fluence rates
also used a higher dose of ALA [7]. PDT caused a 93% no greater than 100 mW/cm2) that we are aware of involved
reduction in mean total, but relative, vascular perfusion of topical administration of ALA to basal cell cancers and squa-
the fibrosarcomas. PDT with ALA has also been reported to mous cell carcinomas in situ [20]. Laser Doppler perfusion
decrease blood flow in normal cremaster muscle of rats [12]. imaging of these lesions showed an increase in perfusion
Microscopic vasoconstriction has been observed in ALA- immediately following PDT. It is not clear if these differing
treated rats with transplanted mammary tumor 5 min after results in humans are due to differences in tumors, fluence
PDT [27]. ‘Partial disruption’ of Colo26 tumor perfusion in rates, fluences, penetration depth of the laser or route of ALA

Thursday Nov 25 01:32 PM StyleTag -- Journal: JPB (J. Photochem. Photobiol. B: Biol.) Article: 7874
104 M.A. Herman et al. / J. Photochem. Photobiol. B: Biol. 52 (1999) 99–104

administration. Nevertheless, the present observations on levels of protoporphyrin in vivo, J. Photochem. Photobiol. B: Biol. 40
blood flow suggest that effects of ALA-based PDT on ade- (1997) 107–110.
[15] J. Webber, D. Kessel, D. Fromm, On-line fluorescence of human
nocarcinomas of the colon and rectum as well as an intra- tissues after oral administration of 5-aminolevulinic acid, J. Photo-
abdominal carcinoid tumor in humans is more pronounced chem. Photobiol. B: Biol. 38 (1997) 209–214.
than would be predicted by some animal studies. [16] J.C.M. Bremner, J.K. Bradley, I.J. Stratford, G.E. Adams, Magnetic
resonance spectroscopic studies on ‘real-time’ changes in RIF-1 tum-
our metabolism and blood flow during and after photodynamic ther-
References apy, Br. J. Cancer 69 (1993) 1083–1087.
[17] J. Mattiello, J.L. Evelhoch, E. Brown, A.P. Schaap, F.W. Hetzel,
[1] S.H. Selman, R.W. Keck, J.E. Klaunig, M. Kreimer-Birnbaum, P.J. Effect of photodynamic therapy on RIF-1 tumor metabolism and blood
Goldblatt, S.L. Britton, Acute blood flow changes in transplantable flow examined by 31P and 2H NMR spectroscopy, NMR in Biomedi-
FANFT-induced urothelial tumors treated with hematoporphyrin cine 3 (1990) 64–70.
derivative and light, Surg. Forum 34 (1983) 676–678. [18] U. Schmidt-Erfurth, T.J. Flotte, E.S. Gragoudas, et al., Benzopor-
[2] S.H. Selman, M. Kreimer-Birnbaum, J.E. Klaunig, P.J. Goldblatt, phyrin-lipoprotein-mediated photodestruction of intraocular tumors,
R.W. Keck, S.L. Britton, Blood flow in transplantable bladder tumors Exp. Eye Res. 62 (1996) 1–10.
treated with hematoporphyrin derivative and light, Cancer Res. 44 [19] P. Sieg, J. Rosperich, A. Alther, Laser blood-flow measurements in
(1984) 1924–1927. malignant tumors during photodynamic therapy (PDT) — an exper-
[3] W.M. Star, H.P.A. Marijnissen, A.E. van den Berg-Blok, J.A.C. imental study, Int. J. Oral Maxillofac. Surg. 23 (1994) 437–439.
Versteeg, K.A.P. Franken, H.S. Reinhold, Destruction of rat mammary [20] I. Wang, S. Andersson-Engels, G.E. Nilsson, K. Wardell, K. Svanberg,
tumor and normal tissue microcirculation by hematoporphyrin deriv- Superficial blood flow following photodynamic therapy of malignant
ative photoradiation observed in vivo in sandwich observation cham- and non-melanoma skin tumours measured by laser Doppler perfusion
bers, Cancer Res. 46 (1986) 2532–2540. imaging, Br. J. Dermatol. 136 (1997) 184–189.
[4] T.J. Wieman, T.S. Mang, V.H. Fingar, T.G. Hill, M.W.R. Reed, T.S. [21] Z. Chen, T.E. Milner, X. Wang, S. Srinivas, J.S. Nelson, Optical
Corey, V.Q. Nguyen, E.R. Render, Effect of photodynamic therapy Doppler tomography: imaging in vivo blood flow dynamics following
on blood flow in normal and tumor vessels, Surgery 104 (1988) 512– pharmacological intervention and photodynamic therapy, Photochem.
517. Photobiol. 67 (1998) 56–60.
[5] B.J. Tromberg, A. Orenstein, S. Kimel, S.J. Barker, J. Hyatt, J.S. [22] K. Johansson, H. Ahn, J. Lindhagen, O. Lundgren, Tissue penetration
Nelson, M.W. Berns, In vivo tumor oxygen tension measurements for and measuring depth of laser Doppler flowmetry in the gastrointestinal
the evaluation of the efficiency of photodynamic therapy, Photochem. application, Scand. J. Gastroenterol. 22 (1987) 1081–1088.
Photobiol. 52 (1990) 375–385. [23] T. Iwao, A. Toyonaga, M. Ikegami, K. Oho, M. Sumino, M. Sakaki,
[6] I.P.J. van Geel, Y.G. Oppelaar, Y.G. Oussoren, F.A. Stewart, Changes H. Harada, K. Tanikawa, Observer agreement and variability in meas-
in perfusion of mouse tumours after photodynamic therapy, Int. J. uring gastric mucosal blood flow by laser Doppler flowmetry in
Cancer 56 (1994) 224–228. humans, Endoscopy 25 (1993) 274–277.
[7] D.J.H. Roberts, F. Cairnduff, I. Driver, B. Dixon, S.B. Brown, Tumor [24] M. Stucker, A. Heese, K. Hoffmann, A. Rochling, P. Altmeyer, Pre-
vascular shutdown following photodynamic therapy based on poly- cision of laser Doppler scanning in clinical use, Clin. Exper. Dermat.
haematoporphyrin or 5-aminolevulinic acid, Int. J. Oncol. 5 (1994) 20 (1995) 371–376.
763–768. [25] L. Bungum, S. Kullander, J.M. Maltau, Laser Doppler flowmetry of
[8] V.H. Fingar, Vascular effects of photodynamic therapy, J. Clin. Laser human endometrial microvasculature — a preliminary communica-
Med. & Surg. 14 (1996) 323–328. tion, Acta Obstet. Gynecol. Scand. 75 (1996) 178–182.
[9] J. Regula, A.J. MacRobert, A. Gorchein, G.A. Buonaccorsi, S.M. [26] M.K. Schilling, C. Redaelli, C. Maurer, H. Friess, M.W. Buchler,
Thorpe, G.M. Spencer, A.R.W. Hatfield, S.G. Bown, Photosensitiza- Gastric microcirculatory changes during gastric tube formation:
tion and photodynamic therapy of oesophageal, duodenal, and colo- assessment with laser Doppler flowmetry, J. Surg. Res. 62 (1996)
rectal tumors using 5-aminolaevulinic acid induced protoporphyrin 125–129.
IX — a pilot study, Gut 36 (1995) 67–75. [27] N. van der Veen, H.L.L.M. van Lengoed, W.M. Star, In vivo fluores-
[10] Q. Peng, K. Berg, J. Moan, M. Kongshaug, J.M. Nesland, 5-Amino- cence kinetics and photodynamic therapy using 5-aminolaevulinic
levulinic acid-based photodynamic therapy: principles and experi- acid-induced porphyrin: increased damage after multiple irradiations,
mental research, Photochem. Photobiol. 65 (1997) 235–251. Br. J. Cancer 70 (1994) 867–872.
[11] J. Webber, D. Kessel, D. Fromm, Side effects and photosensitization [28] B.W. Henderson, L Vaughan, D.A. Bellner, H. van Leengoed, P.G.
of human tissues after aminolevulinic acid, J. Surg. Res. 68 (1997) Johnson, A.R. Oseroff, Photosensitization of murine tumor, vascula-
31–37. ture and skin by 5-aminolevulinic acid-induced porphyrin, Photo-
[12] J. Leveckis, N.J. Brown, M.W.R. Reed, The effect of aminolevulinic chem. Photobiol. 62 (1995) 780–789.
acid-induced, protoporphyrin IX-mediated photodynamic therapy on [29] J. Webber, Y. Luo, R. Crilly, D. Fromm, D. Kessel, An apoptotic
the cremaster muscle microcirculation in vivo, Br. J. Cancer 72 response to photodynamic therapy with endogenous protoporphyrin
(1995) 1113–1119. in vivo, J. Photochem. Photobiol. B: Biol. 35 (1996) 209–211.
[13] J. Bedwell, A.J. MacRobert, D. Phillips, S.G. Bown, Fluorescence [30] A. Orenstein, G. Kostenich, L. Roitman, Y. Shectman, Y. Kopolovic,
distribution and photodynamic effect of ALA-induced PPIX in the B. Ehrenberg, Z. Malik, A comparative study of tissue distribution
DMH rat colonic tumour model, Br. J. Cancer 65 (1992) 818–824. and photodynamic therapy selectivity of chlorin e6, Photofrin II and
[14] M.A. Herman, J. Webber, Y. Luo, V. Patacsil, D. Kessel, D. Fromm, ALA-induced protoporphyrin IX in a colon carcinoma model, Br. J.
Effects of fractionated 5-aminolevulinic acid administration on tissue Cancer 73 (1996) 937–944.

Thursday Nov 25 01:32 PM StyleTag -- Journal: JPB (J. Photochem. Photobiol. B: Biol.) Article: 7874

You might also like