You are on page 1of 7

Current Perspective

Photoangioplasty
An Emerging Clinical Cardiovascular Role for Photodynamic Therapy
Stanley G. Rockson, MD; David P. Lorenz, MD; Wai-Fung Cheong, PhD; Kathryn W. Woodburn, PhD

AbstractPhotodynamic therapy (PDT) has been studied and applied to various disease processes. The potential of PDT
for selective destruction of target tissues is especially appealing in cardiovascular disease, in which other existing
interventional tools are somewhat nonselective and carry substantial risk of damage to the normal arterial wall.
Enthusiasm for photoangioplasty (PDT of vascular de novo atherosclerotic and, potentially, restenotic lesions) is fueled
by more effective second-generation photosensitizers and technological advances in endovascular light delivery. This
excitement revolves around at least 4 significant attributes of light-activated therapy: the putative selectivity and safety
of photoangioplasty, the potential for atraumatic and effective debulking of atheromatous plaque through a biological
mechanism, the postulated capability to reduce or inhibit restenosis, and the potential to treat long segments of abnormal
vessel by simply using fibers with longer light-emitting regions. The available nonclinical data, coupled with the
observations of a new phase I trial in human peripheral atherosclerosis, suggest a promising future for photoangioplasty
in the treatment of primary atherosclerosis and prevention of restenosis. (Circulation. 2000;102:591-596.)
Downloaded from http://circ.ahajournals.org/ by guest on March 21, 2017

Key Words: photodynamic therapy photoangioplasty atherosclerosis restenosis

P hotodynamic therapy (PDT) is an emergent therapeutic


modality for several disease processes. The salutary
response is mediated by a photosensitizing drug, typically
ant photobiological response (direct, rapid cell apoptosis and
delayed necrosis from neovascular damage) becomes maxi-
mal within several days.1,57 Throughout this process, the
administered parenterally and selectively absorbed or re- drug serves simply as a catalyst for energy transfer. Light
tained within the tissues targeted for therapy.1 Differential absorption leads to the release of cytotoxic singlet oxygen, a
selectivity or retention promotes selective damage when the highly reactive, oxidizing agent with very short diffusion
target tissue is exposed to light of an appropriate wavelength; distances (0.1 m). Cell death is thus confined to those
the surrounding normal tissue, containing little or no drug, illuminated areas in which there is an adequate presence of
absorbs little light and is thus spared injury.1,2 Selectivity the sensitizing drug.8
renders PDT particularly appealing in coronary artery dis- The physicochemical properties of the photosensitizing
ease, in which other catheter-based approaches are relatively molecule will predicate preferential uptake by a target cellu-
nonselective and carry a substantial risk of damage to the lar population.5 In atherosclerosis, drug lipophilicity and the
normal arterial wall. Historically, PDT clinical research has high lipid content of vascular plaque both appear to predicate
focused primarily and successfully on treatment of cancer.2 selective uptake. Although these mechanisms have not been
Recent advances in synthetic macrocyclic chemistry, laser established with certainty,4 both passive and active processes
technology, and endovascular light delivery systems have have been identified for the uptake of hydrophobic photosen-
broadened the scope of PDT to include, among others, sitizers into atheromatous plaque.9 Interestingly enough, the
ophthalmic, urologic, immunological, and more recently selective accumulation by atheromatous tissues of some
atherosclerotic applications.3,4 PDT may have a viable role as lipophilic and hydrophilic agents is comparable.10,11 It is
a clinical inhibitor of restenosis and de novo disease. likely therefore that passive diffusion into the arterial wall
from the vasa vasorum and from the lumen, as well as
Mechanism of PDT Action impaired endothelial permeability,12,13 can augment active
The PDT response begins with a differential accumulation of transport mechanisms.
a photosensitizer in the target tissue, the consequence of Whether in neoplasm or in atheroma, light of the appro-
either selective uptake of the drug or of preferential retention priate wavelength is necessary to activate the drug. The light
caused by slower clearance from the abnormal tissue. Subse- can originate from collimated sources (eg, lasers) or from
quently, the tissue is photo-illuminated at the wavelength that diffuse illuminators (eg, high-power lamps and light-emitting
favors maximal absorption by the photosensitizer. The result- diode panels).14,15 The emission wavelength is usually

From the Division of Cardiovascular Medicine, Stanford University School of Medicine, Stanford, Calif (S.G.R., D.P.L.), and Pharmacyclics, Inc,
Sunnyvale, Calif (W.-F.C., K.W.W.).
Correspondence to Stanley G. Rockson, MD, Division of Cardiovascular Medicine, Stanford University School of Medicine, 300 Pasteur Dr, CVRC
287, Stanford, CA 94305. E-mail rockson@leland.stanford.edu
2000 American Heart Association, Inc.
Circulation is available at http://www.circulationaha.org

591
592 Circulation August 1, 2000

matched to the far-red absorption bandwidth of the photosen- molecules, including the phthalocyanines (photoactivated at
sitizer. The light is then delivered to the treatment site either 675 nm),11,33 chlorins (at 660 nm),34 purpurins (at 663 nm),35
directly or via hollow wave guides or fibers terminating in and benzoporphyrin derivatives (at 690 nm).13 However,
optical configurations to achieve either a circumferential, most of these agents require liposomal or intralipid formula-
segmental, or conical beam profile.16 Radially emitting fibers, tion before administration.
with a variety of designs, have been used for endovascular Early PDT agents for cardiovascular disease were activated
PDT in animals,1719 and in one case, a 2-patient clinical at wavelengths 700 nm, at which point blood and tissues
study.17 Another important advance in photoangioplasty is substantially attenuate the delivery of light to target cells.36
the development of compact, portable, and relatively inex- However, tissue optics dictate that for optimal photochemical
pensive diode-based lasers to replace the older, large systems response, the ideal photosensitizer should display maximal
requiring special electrical and plumbing infrastructures; the absorption in the range of 700 to 800 or 950 to 1100 nm.15
latter are mostly dye lasers pumped by another laser (ar- The recent renewal of interest in the therapeutic potential of
gon,19 21 KTP YAG) or pulse lasers (copper vapor).19 cardiovascular PDT has been prompted largely by the avail-
Severe cellular damage is noted in atheromatous areas after ability of expanded macrocycles known as the texaphyrins.
light exposure.7 However, there is demonstrable preservation These drugs circumvent many of the physicochemical limi-
of the intact elastic lamina and of normal collagen in the tations of previously studied sensitizers. Texaphyrins are
adventitia, suggesting that the functional integrity of the synthetic, water-soluble macrocycles with long wavelength-
vessel is conserved.22 The absence of mural inflammation, absorbing properties. They localize both in cancerous lesions
despite extensive cell death, is consistent with the regression and in atheromatous plaque.20,37,38 Incorporation of a diamag-
of atherosclerotic plaque through a hypothesized mechanism
Downloaded from http://circ.ahajournals.org/ by guest on March 21, 2017

netic lanthanide, lutetium, into the texaphyrin molecule yields


of apoptosis, although this has not yet been conclusively a potent PDT agent that is activated by tissue-penetrating
demonstrated. Paradoxically, a so-called dark effect, ie, a far-red light (732 nm).20,21 In addition, motexafin lutetium
therapeutic response to the drug in the absence of light, has fluoresces at 750 nm (Figure 1); endogenous chromophores
also been described for the inhibitory effect on restenosis of do not emit light at 750 nm. Hence, in vivo real-time imaging
at least one photosensitizer, a benzoporphyrin derivative. The of target structures is feasible, thus facilitating clinical diag-
therapeutic implication of this observation has not been nosis and treatment planning. Similarly, the related, paramag-
delineated. netic gadolinium texaphyrin might facilitate MRI of athero-
matous vascular disease.37
Photosensitizing Drugs for PDT Texaphyrins localize in and eradicate diseased tissues,20
Hematoporphyrin derivative (HpD) was the first of a number including atherosclerotic plaque (Figure 2).21,37 PDT with
of photosensitizers with demonstrable, selective accumula- motexafin lutetium causes selective photodamage and
tion within atherosclerotic plaque.23 Subsequent studies have
thereby helps to reverse both diet-induced20 and balloon-
underscored the affinity of porphyrin derivatives for diseased
induced39 atheromatous plaque in rabbits. The enhanced
arterial wall in rabbits24 26 and miniswine.12,13 There is
efficacy of texaphyrins may be attributable both to a more
maximal drug concentration in the intimal surface layers,
selective uptake and retention of the photosensitizing mole-
diminishing radially into the media.27,28 Both hematoporphy-
cules and to the depth of light penetration achievable in blood
rin29 and Photofrin,30 a more purified derivative of HpD, also
and tissue at the longer 732-nm wavelength illumination.20
display in vitro preferential uptake by human plaque. Al-
Uptake of motexafin lutetium by the atheromatous plaque
though Photofrin is available for clinical use in antineoplastic
occurs in a ratio of 16:1 to 34:1 when diseased and normal
applications, its clinical performance in the PDT of athero-
segments of the arterial wall are contrasted.40 One possible
sclerosis has been somewhat disappointing.17 Despite obvi-
mechanism for this selective plaque uptake of texaphyrins is
ous, selective damage of the plaque and sparing of the
through plasma lipoprotein binding or modification.37 Unlike
underlying media, the lack of efficacy might be attributable to
HpD-based PDT,21 there is no evidence of microscopic
inadequate penetration of the 630-nm light through endolu-
damage to the arterial wall after PDT with motexafin
minal blood. Clinical application is further hampered by the
lutetium.
propensity of treated subjects to display prolonged cutaneous
phototoxicity.4
Administration of 5-aminolevulinic acid (ALA), a bio-
Cardiovascular Applications of PDT
chemical precursor for protoporphyrin IX, has been accom- In Vitro Experience
plished by topical, systemic, and local internal routes in a The extent of the short-term angiographic improvement plays
variety of malignant and dysplastic conditions.8,31 However, a determining role in the durability of the clinical response to
its administration can elicit hemodynamic changes (depres- standard, percutaneous vascular interventions; however, re-
sion of systemic and pulmonary pressures and pulmonary stenosis still significantly impairs the potential for long-term
resistance) that might limit its ultimate utility in cardiovas- clinical benefits for many patients.41 The identified biological
cular applications.32 determinants of the restenosis response appear to be, in
Newer agents with selective localization, greater PDT principle, amenable to the mechanisms of phototherapy.
efficiency, and minor, self-limited potential for cutaneous These determinants include vascular remodeling, stimulation
phototoxicity are now available.4 Phototherapeutic capacity of the proliferative and migratory response of vascular
in atherosclerosis has been described for a number of these smooth muscle cells (SMCs), and enhanced production of
Rockson et al Photodynamic Therapy in Cardiovascular Medicine 593

Figure 1. Motexafin lutetium: chemical struc-


ture, absorbance, and fluorescence profiles.

extracellular matrix.39,42 Furthermore, neutrophil and platelet erosclerotic arteries.30,47 The effect on SMCs obtained from
Downloaded from http://circ.ahajournals.org/ by guest on March 21, 2017

activation has been demonstrated with angioplasty in balloon- atherosclerotic lesions (activated SMCs) is much greater than
injured arterial plaque.43 Moreover, macrophages play an that seen in the cells obtained from the normal vascular wall.
important role in the complex activation of SMCs after Similarly, studies of SMCs derived from saphenous vein
vascular injury.44 grafts have shown significant inhibition of their growth after
Several studies have shown that PDT inhibits SMC growth photosensitization.48 In addition, PDT on bovine aortic endo-
and decreases the development of experimentally induced thelial cell preparations in vitro induces changes in extracel-
intimal hyperplasia response. The effects of PDT on the lular matrix: SMC proliferation and migration are inhibited
injury response seem to be rather complex. Photosensitization and endothelial cell proliferation is enhanced.48 These PDT-
can accomplish a complete cellular eradication within the induced vascular responses may benefit the process of vas-
vascular wall without associated inflammation and prolifera- cular remodeling and reduce the likelihood of a restenosis
tion, suggesting that PDT may induce changes in the extra- response. The preferential uptake of the drug in atheroscle-
cellular matrix of the vascular wall.45 In vitro, exposure to rotic segments is further accentuated in the highly cellular
PDT eliminates detectable levels of basic fibroblast growth regions of restenosis, suggesting that the selective cytotoxic
factor (FGF and FGF-2) in solution and significantly re- effect could be applied to both the therapy and the prophy-
duces the smooth muscle mitogenesis inducible by matrix- laxis of restenosis.45
associated FGF-2.45 In vivo, PDT of rat carotid arteries
produces a loss of FGF staining compared with control, In Vivo Experience
nontreated arteries. Furthermore, the effect of PDT on the A benefit after photosensitization with Photofrin in athero-
release and activation of transforming growth factor-1 has sclerotic rabbits has been reported,24,49 although others have
also been examined in vitro.46 The data suggest that PDT may shown only a slight to modest reduction in plaque burden.27
inhibit intimal hyperplasia through local inhibition of local The lack of marked therapeutic benefit with Photofrin-
cytokine release or activation. mediated PDT in atherosclerosis has been ascribed to the
In vitro investigations support the concept that PDT can ability of blood to impair light transmission at 630 nm.12
favorably influence endothelial vascular biology. PDT with Thus, suboptimal light transmission would produce subthera-
Photofrin II substantially impairs the growth of cultured peutic activation of the sensitizer. Furthermore, safety con-
SMCs derived from both atherosclerotic lesions and nonath- siderations limit the maximum light dose that can be deliv-

Figure 2. Spectral bioimaging of


motexafin lutetium within atheromatous
plaque compared with surrounding nor-
mal tissue. Hypercholesterolemic rabbit
received intravenous dose of motexafin
lutetium (10 mol/kg). a, Aortic lumen
was exposed at 24 hours after injection
to fluorescence imaging. b, Spectral
classification confirms selective accumu-
lation and retention of photosensitizer
within atheroma. c, Fluorescence emis-
sion profiles of atheroma and normal
regions denoted in Figure 2a.
594 Circulation August 1, 2000

Figure 3. Three microscopic hematoxylin and eosinstained cross sections of cholesterol-fed rabbit artery obtained 2 weeks after PDT
with motexafin lutetium. Intima is extensively thickened and heavily laden with foam cells. Photoirradiated site (b) demonstrates sub-
stantial reduction of disease burden, while vascular regions above (a) and below (c) treatment zone remain heavily burdened with
plaque.

ered. In experimental treatments of canine coronary arteries intimal hyperplasia but did not prevent it. The authors
in vivo, light doses 200 J/cm2 elicited angiographic spasm, concluded that refinements in dosimetry will be necessary to
histological necrosis, and even transmural injury.18 In this achieve long-term benefits.53
study, the cases of premature death were ascribed to the The importance of the selection of the correct arterial
effects of coronary artery spasm, because only minor medial region in the prevention of restenosis has been examined in
damage was seen in some of these specimens. In fact, even at the balloon-injured rat carotid artery.54 The results of that
high light doses, no embolization, vessel perforation, or study indicate that a hypercellular injury response in a treated
aneurysmal dilatation was seen. lesion can originate from a remote source; consequently,
Downloaded from http://circ.ahajournals.org/ by guest on March 21, 2017

Efficient reduction of atherosclerotic plaque burden has successful elimination of restenosis by PDT may require
also been demonstrated after PDT with motexafin lutetium. inclusion of the entire injured artery in the treatment field,
Histological analysis of the posttreatment specimens reveals perhaps including a section of uninjured margin.
selective reduction in plaque area in a hypercholesterolemic
rabbit model (Figure 3),21 whereas in a balloon-injury model, Human Clinical Applications
a significant reduction in macrophage density within the Photoangioplasty for arterial diseases, including de novo
treated lesions was observed.39 atherosclerosis and potentially restenosis, has reemerged
The in vivo studies on intimal hyperplasia have yielded because of promising new drugs and safer, less expensive
more universally promising results. For example, a substan- optical devices. This approach has the intriguing potential for
tial reduction in intimal hyperplasia has been shown in the rat invoking separate but interrelated mechanisms of benefit
carotid artery injury model with chloroaluminum-sulfonated from one therapeutic intervention: safe debulking of the
phthalocyanine and 675-nm light at a fluence (the power atheroma through selective injury and destruction of the
density of light over time) of 100 J/cm2.50 No thermal injury atherosclerotic material; inhibition of the restenosis process
was identified in the treated vascular segments. In most cases, through an effect on macrophages, intimal hyperplasia, and
inhibition of intimal hyperplasia correlates with histological the inhibition of SMC proliferation; and further inhibition of
absence of inflammatory and SMCs in the media. Similar restenosis through a hypothesized dark effect of some
benefits have been observed in other experimental model photosensitizers.4
systems with Photofrin51 and ALA.8 PDT at the time of The clinical trials of motexafin lutetium (Antrin) were
angioplasty leads to an acellular media despite regeneration propelled by early preclinical indications of selective and
of the endothelial lining.8 On the basis of the latter study, it efficacious resolution of plaque in rabbits. Clinical evaluation
would appear that it is not necessary to delay PDT after of motexafin lutetium is also ongoing in patients with
balloon injury to prevent the injury response of restenosis, recurrent breast cancer (Lutrin) and age-related macular
because photoactivation at the time of angioplasty completely degeneration (Optrin). In these latter studies, the drug has
abolished the expected intimal hyperplasia after this injury. In been well tolerated. The maximum tolerated dose proved to
addition, both short- and long-term benefits of PDT have be 5.5 mg/kg on the basis of elicitation of pain in the
been demonstrated.45,52 treatment field and dysesthesias in light-exposed areas.55
In another study of PDT in a balloon-injured rodent arterial Plasma pharmacokinetic data taken from these patients
model, the media remained acellular for several weeks to showed the drug to be cleared relatively quickly, exhibiting a
months, and intimal hyperplasia did not occur.6 Although T1/2 and T1/2 of 0.32 and 12.9 hours, respectively. Early
endothelial regeneration occurred by 2 weeks, SMCs failed to observations from a phase I trial in claudicants with periph-
repopulate the media. During PDT, retraction of endothelial eral arterial atherosclerosis suggest that the therapy is well
cells does allow adherence of neutrophils by their 2-integrin tolerated and has the capacity to invoke a therapeutic re-
adhesion receptors to the subendothelial matrix, leading to the sponse in these patients. In 90% of vessels treated to date,
hypothesis that successful prevention of intimal hyperplasia intravascular ultrasonography has confirmed measurable im-
by PDT relies in part on the presence of the neutrophil at the provement in lumen cross-sectional area after Antrin pho-
site of the lesion. Recently, Photofrin PDT was performed toangioplasty. The therapeutic changes are achieved without
with continuous external laser irradiation in the rabbits either documented adverse vascular responses or any treatment-
1 (prevention) or 6 (treatment) weeks after balloon injury.53 limiting phototoxicity. In these ongoing trials, doses of 1 to 5
PDT was quite effective in the treatment of established mg/kg of Antrin have been administered intravenously before
Rockson et al Photodynamic Therapy in Cardiovascular Medicine 595

PDT, although preclinical data also support the feasibility of 14. Wilson B. Light sources for photodynamic therapy. Photodynamic News.
local endovascular drug delivery. This may ultimately be 1998;1:6 8.
15. Doiron D. Photophysics and instrumentation for porphyrin detection and
more clinically advantageous.39,56 Endovascular light is de- activation. In: Doiron C, Gomer C, eds. Porphyrin Localization and
livered through a cylindrical diffuser fiber, typically 24 hours Treatment of Tumors. New York, NY: Liss; 1984:4173.
after systemic, intravenous administration of Antrin. The 16. Van den Berg H. On the evolution of some endoscopic light delivery
fiber is positioned adjacent to the lesion of interest by systems for photodynamic therapy. Endoscopy. 1998;30:393 407.
17. Spears J. Photosensitization. In: Isner J, Clarke R, eds. Cardiovascular
percutaneous delivery through a standard 5F to 8F guiding
Laser Therapy. New York NY: Raven Press Ltd; 1989:107120.
catheter. Light treatment is sustained for 941 seconds to 18. Mackie RW, Vincent GM, Fox J, et al. In-vivo canine coronary artery
achieve 400-J/cm diffuser fiber over a 3-cm diffuser length laser irradiation: photodynamic therapy using dihematoporphyrin ether
fiber. A portable, relatively inexpensive 730-nm diode system and 632 nm laser. Lasers Surg Med. 1991;11:535544.
facilitates these illumination requirements. 19. Sobeh M, Greenwald S, Ham R, et al. Induction or prevention of intimal
hyperplasia by photodynamic therapy in the porcine model. SPIE. 1995;
Of paramount interest may be the capacity of photoangio- 2395:390 395.
plasty to prevent the cellular responses of restenosis after 20. Young SW, Woodburn KW, Wright M, et al. Lutetium texaphyrin (PCI-
conventional endovascular procedures. Future investigation 0123): a near-infrared, water-soluble photosensitizer. Photochem Pho-
will also determine the synergistic role of photoangioplasty tobiol. 1996;63:892 897.
21. Woodburn K, Fan Q, Kessel D, et al. Phototherapy of cancer and ather-
when performed at the time of standard endovascular proce- omatous plaque with texaphyrins. J Clin Laser Med Surg. 1996;14:
dures. The available preclinical investigations, coupled with 343348.
recent and continuing improvements in laser and fiberoptic 22. Grant W, Speight P, MacRobert A, et al. Photodynamic therapy of normal
technology, suggest a promising role for photoangioplasty in rat arteries after photosensitisation using disulphonated aluminum phtha-
locyanine and 5-aminolaevulinic acid. Br J Cancer. 1994;70:7278.
the future prevention and treatment of restenosis. Further
Downloaded from http://circ.ahajournals.org/ by guest on March 21, 2017

23. Spears J, Serur J, Shropshire D, et al. Fluorescence of experimental


clinical investigation must evaluate therapeutic outcomes and atheromatous plaques with hematoporphyrin derivative. J Clin Invest.
exclude a significant potential for coronary artery spasm in 1983;71:395399.
human applications. Although the time needed for full cyto- 24. Neave V, Gianotta S, Hyman S, et al. Hematoporphyrin uptake in ath-
toxic effect may limit applicability to short-term, primary erosclerotic plaques: therapeutic potentials. Neurosurgery. 1988;23:
307312.
therapeutic interventions, one can envision a potential role of 25. Pollock M, Eugene J, Hammer-Wilson M, et al. Photosensitization of
photoangioplasty as an adjunct to the standard percutaneous experimental atheromas by porphyrins. J Am Coll Cardiol. 1987;9:
techniques for revascularization. Additional, ripe clinical 639 646.
scenarios might include interventions for vulnerable plaque, 26. Pernes JM, Brault D, Angel CY, et al. Comparative study of the selective
uptake of hematoporphyrin derivative components into experimental ath-
long coronary lesions, diffuse and distal vascular disease, and
eromatous plaques: potential for laser angioplasty. J Intervent Radiol.
stabilization of vulnerable plaque. 1988;3:3 8.
27. Litvack F, Grundfest WS, Forrester JS, et al. Effects of hematoporphyrin
References derivative and photodynamic therapy on atherosclerotic rabbits. Am J
1. Henderson B, Dougherty T. How does photodynamic therapy work? Cardiol. 1985;56:667 671.
Photochem Photobiol. 1992;55:145157. 28. Pagnan A, Scannapieco G, Pauletto P, et al. Haematoporphyrin and
2. Dougherty T. Photosensitizers: therapy and detection of malignant atherosclerosis in the broad-breasted white turkey: distribution and quan-
tumors. Photochem Photobiol. 1987;45:879 889. titation in thoracic and abdominal aorta. Int J Tissue React. 1989;11:
3. Levy J. Photodynamic therapy. Trends Biotechnol. 1995;13:14 18. 9399.
4. Vincent G. Photodynamic therapy of atherosclerosis and restenosis: a 29. Kessel D, Sykes E. Porphyrin accumulation by atheromatous plaques of
potentially exciting new treatment method. SPIE. 1994;2130:210. the aorta. Photochem Photobiol. 1984;40:59 61.
5. Kessel D, Luo Y, Deng Y, et al. The role of subcellular localization in 30. Dartsch P, Ischinger T, Betz E. Responses of cultured smooth muscle
initiation of apoptosis by photodynamic therapy. Photochem Photobiol. cells from human nonatherosclerotic arteries and primary stenosing
1997;65:422 426. lesions after photoradiation: implication for photodynamic therapy of
6. Sluiter W, de Vree W, Pietersma A, et al. Prevention of late lumen loss vascular stenosis. J Am Coll Cardiol. 1990;15:15451550.
after coronary angioplasty by photodynamic therapy: role of activated 31. Kennedy J, Marcus S, Pottier R. Photodynamic therapy (PDT) and pho-
neutrophils. Mol Cell Biochem. 1996;157:233238. todiagnosis (PD) using endogenous photosensitization induced by
7. Reed M, Miller F, Wieman T, et al. The effect of photodynamic therapy 5-aminolevulinic acid (ALA): mechanisms and clinical results. J Clin
on the microcirculation. J Surg Res. 1988;45:452 459. Laser Med Surg. 1996;14:289 304.
8. Nyamekye I, Anglin S, McEwan J, et al. Photodynamic therapy of normal 32. Herman M, Webber J, Fromm D, et al. Hemodynamic effects of
and balloon-injured rat carotid arteries using 5-amino-levulinic acid. 5-aminolevulinic acid in humans. J Photochem Photobiol B. 1998;43:
Circulation. 1995;91:417 425.
61 65.
9. Delettre E, Brault D, Bruneval P, et al. In vitro uptake of dicarboxylic
33. LaMuraglia G, Ortu P, Flotte T, et al. Chloroaluminum sulfonated phtha-
porphyrins by human atheroma: kinetic and analytical studies. Photochem
locyanine partitioning in normal and intimal hyperplastic artery in the rat:
Photobiol. 1991;54:239 246.
implications for photodynamic therapy. Am J Pathol. 1993;142:
10. Straight R, Vincent G, Hammond E, et al. Porphyrin retention and
1898 1905.
photodynamic treatment of diet induced atherosclerotic lesions in pigs. In:
Jori G, Perria C, eds. Photodynamic Therapy of Tumors and Other 34. Hayashi J, Kuroiwa Y, Sato H, et al. Transadventitial localization of
Diseases. Padova, Italy: Libreria Progetto; 1986:350 352. atheromatous plaques by fluorescence emission spectrum analysis of
11. Eldar M, Yerushalmi Y, Kessler E, et al. Preferential uptake of a water mono-L-aspartyl chlorin e6. Card Res. 1993;27:19431947.
soluble phthalocyanin by atherosclerotic plaques in rabbits. Atheroscle- 35. Narciso, H, Anderson S, DeHoratius S, et al. SnET2 for the treatment of
rosis. 1990;84:135139. vascular disease: dose/response study in NZW rabbits. SPIE. 1995:2395:
12. Vincent G, Mackie R, Orme E, et al. In vivo photosensitizer enhanced 409 417.
laser angioplasty in atherosclerotic Yucatan miniswine. J Clin Laser Med 36. Vincent G, Fox J, Charlton G, et al. Presence of blood significantly
Surg. 1990;8:59 61. decreases transmission of 630 nm laser light. Lasers Surg Med. 1991;11:
13. Hsiang Y, Crespo M, Richter A, et al. In vitro and in vivo uptake of 399 403.
benzoporphyrin derivative into human and miniswine atherosclerotic 37. Woodburn K, Young S, Fan Q, et al. Selective uptake of texaphyrins by
plaque. Photochem Photobiol. 1993;57:670 674. atheromatous plaque. SPIE. 1996;2671:6271.
596 Circulation August 1, 2000

38. Sessler J, Hemmi G, Mody T, et al. Texaphyrins: synthesis and appli- 48. Adili F, Statius van Eps R, Karp S, et al. Differential modulation of
cations. Acc Chem Res. 1994;27:4350. vascular endothelial and smooth muscle cell function by photodynamic
39. Miller M, Kuntz R, Friedrich S, et al. Frequency and consequences of therapy of extracellular matrix: novel insights into radical-mediated pre-
intimal hyperplasia in specimens retrieved by directional atherectomy of vention of intimal hyperplasia. J Vasc Surg. 1996;23:698 705.
native primary coronary artery stenoses and subsequent restenosis. Am J 49. Tang G, Hyman S, Schneider J, et al. Application of photodynamic
Cardiol. 1993;71:652 658. therapy to the treatment of atherosclerotic plaques. Neurosurgery. 1993;
40. Woodburn K, Qing F, Kessel D, et al. Photoeradication and imaging of 32:438 443.
atheromatous plaque with texaphyrins. SPIE. 1997;2970:44 50. 50. Ortu P, LaMuraglia G, Roberts G, et al. Photodynamic therapy of arteries:
41. Serruys PW, Luijten HE, Beatt KJ, et al. Incidence of restenosis after a novel approach for treatment of experimental intimal hyperplasia. Cir-
successful coronary angioplasty: time-related phenomenon: a quantitative culation. 1992;85:1189 1196.
angiographic study in 342 consecutive patients at 1, 2,3, and 4 months. 51. Eton D, Colburn M, Shim V, et al. Inhibition of intimal hyperplasia by
Circulation. 1988;77:361371. photodynamic therapy using Photofrin. J Surg Res. 1992;53:558 562.
42. Simons M, Leclerc G, Safian R, et al. Relation between activated smooth-
52. Gonschior P, Gerheuser F, Fleuchaus M, et al. Local photodynamic
muscle cells in coronary-artery lesions and restenosis after atherectomy.
therapy reduces tissue hyperplasia in an experimental restenosis model.
N Engl J Med. 1993;328:608 613.
Photochem Photobiol. 1996;64:758 763.
43. Neumann F, Ott I, Gawaz M, et al. Neutrophil and platelet activation at
53. Eton D, Shim V, Maibenco TA, et al. Cytotoxic effect of photodynamic
balloon-injured coronary artery plaque in patients undergoing angio-
plasty. J Am Coll Cardiol. 1996;27:819 824. therapy with Photofrin II on intimal hyperplasia. Ann Vasc Surg. 1996;
44. Hanke H, Hassenstein S, Ulmer A, et al. Accumulation of macrophages 10:273282.
in the arterial vessel wall following experimental balloon angioplasty. Eur 54. Statius van Eps R, ChandraSekar N, Hasan T, et al. Importance of the
Heart J. 1994;15:691 698. treatment field for the application of vascular photodynamic therapy to
45. LaMuraglia G, Adili F, Karp S, et al. Photodynamic therapy inactivates inhibit intimal hyperplasia. Photochem Photobiol. 1998;67:337342.
extracellular matrix-basic fibroblast growth factor: insights to its effect on 55. Renschler MF, Yuen AR, Panella TJ, et al. Photodynamic therapy trials
the vascular wall. J Vasc Surg. 1997;26:294 301. with motexafin lutetium (Lu-Tex) in patients with locally recurrent breast
46. Statius van Eps R, LaMuraglia G. Photodynamic therapy inhibits trans- cancer. SPIE. 1998;3247:3539.
Downloaded from http://circ.ahajournals.org/ by guest on March 21, 2017

forming growth factor beta. J Vasc Surg. 1997;25:1044 1052. 56. Gonschior P, Erdemci F, Gerheuser F, et al. Selective hematoporphyrin
47. Dartsch P, Ischinger T, Betz E. Differential effect of Photofrin II on (HMD) application in arterial vessels using a porous balloon catheter
growth of human smooth muscle cells from non atherosclerotic arteries results in equivalent levels as compared to high-dose systemic adminis-
and atheromatous plaques in vitro. Atherosclerosis. 1990;10:616 624. tration [in German]. Z Kardiol. 1991;80:738 745.
Photoangioplasty: An Emerging Clinical Cardiovascular Role for Photodynamic Therapy
Stanley G. Rockson, David P. Lorenz, Wai-Fung Cheong and Kathryn W. Woodburn

Circulation. 2000;102:591-596
doi: 10.1161/01.CIR.102.5.591
Circulation is published by the American Heart Association, 7272 Greenville Avenue, Dallas, TX 75231
Downloaded from http://circ.ahajournals.org/ by guest on March 21, 2017

Copyright 2000 American Heart Association, Inc. All rights reserved.


Print ISSN: 0009-7322. Online ISSN: 1524-4539

The online version of this article, along with updated information and services, is located on the
World Wide Web at:
http://circ.ahajournals.org/content/102/5/591

Permissions: Requests for permissions to reproduce figures, tables, or portions of articles originally published
in Circulation can be obtained via RightsLink, a service of the Copyright Clearance Center, not the Editorial
Office. Once the online version of the published article for which permission is being requested is located,
click Request Permissions in the middle column of the Web page under Services. Further information about
this process is available in the Permissions and Rights Question and Answer document.

Reprints: Information about reprints can be found online at:


http://www.lww.com/reprints

Subscriptions: Information about subscribing to Circulation is online at:


http://circ.ahajournals.org//subscriptions/

You might also like