Treatment of malignant gliomas of the brain remains a serious problem on a global scale, despite intensive research into the causes and mechanisms of their progression. When using traditional surgical approaches and imaging methods tumor cell infiltrates may be overlooked, as a result of which, malignant gliomas relapse often occurs near the marginal region of the surgical cavity. A method that allows visual identification of tumor tissue and at the same time provides an opportunity to selectively destroy it is photodynamic therapy (PDT) ‒ a two-stage treatment that includes the introduction (intravenous, intraperitoneal, local or oral) of a light-sensitive chemical agent (photosensitizer (PS)) followed by its activation at a certain wavelength of light. The principle of PDT is based on the cytotoxic efects caused by PS, which selectively accumulates in malignant tumor cells and is activated by light rays of the appropriate wavelength, generating singlet oxygen and free radicals, which trigger photochemical reactions in tumor cells with subsequent destruction of protein structures. Tumor tissue has a higher afinity for PSs. PSs are divided into 1st, 2nd and 3rd generation molecules. So far, 3rd generation PSs have not yet been approved for clinical use. In vitro and in vivo experimental studies confirmed the efectiveness of PDT of brain tumors using 2nd generation PSs.
Malignant neoplasms are a global medical and
social problem with a tendency to a steadily increasing
morbidity and mortality rates (primarily due to malignant
gliomas (MG)) [
1
]. Treatment of malignant brain gliomas
remains a great challenge on a global scale, despite
intensive research into the causes and mechanisms of
their progression.
Etiology and epidemiology of malignant brain gliomas
Gliomas are conditionally divided into two broad
groups [
2
]: circumscribed gliomas, which have relatively
well-defined margins and can be completely removed
surgically, and difuse gliomas, which have no clear
borders and cannot be totally removed during surgery.
In 2021, the classification of central nervous system
(CNS) tumors was revised taking into account molecular
biological markers (5th edition) [
3
]. According to the
new edition, difuse gliomas in adults are classified into
three groups: astrocytomas (IDH mutation, CNS WHO
grade 2, 3, 4), oligodendrogliomas (IDH mutation and
1p19q codeletion, CNS WHO grade 2, 3), glioblastomas
(IDH-wildtype, CNS WHO grade 4), in children
into low-grade and high-grade with corresponding
subtypes. Circumscribed astrocytic gliomas include The resistance of GB in response to curative
pilocytic astrocytomas, high-grade astrocytomas with measures is widely known and can be explained by
piloid features, pleomorphic xanthoastrocytomas, several features: GB is a heterogeneous population
subependymal giant cell astrocytomas, chordoid with a large number of signaling pathways even within
gliomas, astroblastomas (MN1-altered) [
3
]. a single tumor, which limits the possibilities of targeted
According to CBTRUS statistical report of for therapy; the microenvironment of GB enhances their
2014‒2018 (the previous edition of the CNS tumor resistance to radiation and chemotherapy; the low
classification of 2016 was applied), the average annual immunogenicity of GB cells prevents an adequate
incidence rate of malignant brain tumors and other CNS immunological response; invasion of the perifocal
tumors was 7.06 per 100,000 population, the average zone of the brain by glioma cells (including stem cells)
annual death rate was 4.43 [
2, 4
]. The most common significantly complicates efective treatment by surgical
malignancy among brain tumors and other CNS tumors resection alone; the blood-brain barrier prevents the
is glioblastoma (GB) (14.3% of all tumors and 49.1% of achievement of a suficient concentration in the brain of
malignant brain tumors). The five-year relative survival systemically administered chemotherapy drugs without
rate after morphological diagnosis of a malignant brain serious side efects [
9
]. There is an active search for
tumor and other CNS tumors was 66.9%, whereas for alternative ways of diagnosis and treatment of GB in
non-malignant variants of brain tumors and other CNS the following areas: new technologies of cellular and
tumors it was 92.1%. Difuse gliomas had a significantly molecular diagnostics, nanotechnology, the use of
worse prognosis than circumscribed/demarcated stem cells as carriers of ligands and vectors, modern
gliomas, and differed significantly in the duration approaches to immunotherapy, technologies of targeted
of overall and recurrence-free survival of patients and personalized therapy, etc.
depending on the histological variant [
2
]. Among the most recent approaches are innovative
The incidence of malignant variants of primary intraoperative techniques and various methods of
brain tumors in Ukraine, according to the latest data of treating the "tumor" surgical site. In particular, the
the National Cancer Registry (2020), is 5.0 per 100,000 following options for the introduction of pharmacological
population, the mortality rate is 3.5 per 100,000 population agents are ofered: stereotaxic injections of various
[
5
]. Among primary CNS tumors, malignant forms of brain compounds directly into the tumor using neuronavigation,
gliomas (WHO grade 3-4) predominate, most of them as well as in combination with intraoperative computed
are GB (WHO grade 4). The survival analysis of patients tomography or magnetic resonance imaging (MRI)
with oligoastrocytomas (163 cases for the period from (chemotherapeutic agents, radioactive iodine, iron
2005 to 2015) showed that the median overall survival oxide nanoparticles); convection-enhanced delivery of
was (49.9±2.4) months and difered depending on the compounds using pressure gradient to improve their
histostructure of the tumor: in patients with predominance distribution (chemotherapeutic agents, viral vectors,
of the oligodendroglial pool, the average overall survival of cytotoxins, radioactive nanoliposomes); implantation of
patients who underwent surgery was (100.5±4.6) months, Ommaya or Rickham reservoirs for periodic therapeutic
with predominance of the progression of the astrocytic injections over a long period (for chemotherapy,
component of the proliferate - (48.2±4.5) months, with administration of neural stem cells, cell therapy of
approximately equal distribution of the oligodendroglial CAR (chimaeric antigen receptor) T-cells; intra-arterial
and astrocytic components - (76.6± 4.9) months [
6
]. The delivery of high doses of therapeutic agents (antibodies,
survival analysis of patients with primary GB in Ukraine chemotherapeutic agents, viral vectors) using catheters
(3763 cases for the period from 2008 to 2016) showed inserted directly in the arteries involved in the blood
that the median overall survival was (12.2±0.2) months, supply of the tumor focus [
9
].
the average overall survival of operated patients was The treatment of MG by alternating electric fields,
(7.5±0 ,3) months, when using combined therapy (surgical methods of laser-induced interstitial thermotherapy,
removal, radiation and chemotherapy) – (16.3±0.5) magnetic hyperthermic therapy, focused ultrasound,
months (p<0.001) [
7
]. radiofrequency microwave and photodynamic therapy
Consequently, the results of complex treatment of (PDT) are suggested [
9, 10
].
GB remain unsatisfactory due to the invasive nature of Photodynamic methods, such as photodynamic
their spread and multiresistance to adjuvant treatment diagnosis, f luorescence-guided tumor resec tion
methods and require the development of new and (fluorescence-guided surgery (FGS)), and PDT, are
improvement of existing methods for the management being intensively developed in clinical trials as adjuvant
of this pathology. The modern standard of treatment methods of therapy for MG [
11, 12
].
for GB involves surgical treatment with maximum Invasive glial tumor spread and the degree of
functionally possible resection radiochemotherapy and surgical resection are dificult to definitively assess
adjuvant chemotherapy [
8
]. However, despite advances during surgery, and areas of tumor cell invasion may be
in neurosurgery, chemotherapy, and radiotherapy, overlooked when using traditional surgical approaches
GB remains one of the most treatment-resistant and imaging techniques. In addition, the functional load
CNS malignant neoplasms and the tumor inevitably of certain anatomical areas limits the neurosurgeon
progresses. Most cases of continued growth/recurrence in the radicality of the removal, therefore, MG often
occur in or near the resection area, namely the area that progressively occur near the marginal area of the surgical
received the highest dose of postoperative radiation [
9
]. cavity as a result of residual glioma cells. A method
Many new therapies are designed to combat these local that enables visual identification of tumor tissue while
recurrences by administering therapeutic agents directly selectively destroying it will contribute to better surgical
into or near the tumor bed. removal of MG. Such method is PDT - a two-stage
treatment involving the administration (intravenously,
intraperitoneally, locally or orally) of a light-sensitive
agent (photosensitizer (PS)) followed by its activation
by light rays of a certain wavelength for target lesion
of tumor cells.
History of photodynamic therapy
The beginning of using the energy of photochemical
reactions is associated with Ancient Egypt, when in the
6th millennium BC vitiligo skin disease was treated with
a powder of leaves of parsnip, parsley and
St.-John'swort, which was spread on the afected skin areas, and
when exposed to sunlight, light-sensitive components
contained in these plants caused a photochemical
reaction in tissues, resulting in the appearance of
skin pigmentation [
13
]. Photodynamic procedures are
described in ancient books - the Ebers papyrus and
the sacred Indian book "Atharvaveda", similar methods
(oxygen-free photochemical reactions) were also
practiced in ancient China, Thailand, Arab countries (in
particular, the use of Aatrillal seed powder (later name
- Ammi majus, Chinese cumin)) [
14
].
Over the past 100 years, the role of photosensitizing
agents in the implementation of the antitumor response
to photoirradiation has been investigated. In 1900,
O. Raab in the laboratory of H. von Tappeiner at the
University of Munich established an oxygen-dependent
photodynamic efect of a number of natural dyes (in
particular, acridine orange) during the solar irradiation
of simple unicellular organisms Paramecium [
14, 15
].
H. Jesionek et al. observed an improvement in the
skin condition of patients with tuberculosis and syphilis
after using an aqueous eosin solution and solar rays
or an arc lamp irradiation. H. von Tappeiner and H.
Jesionek (1903) first performed PDT in a skin cancer
patient using eosin solution as a PS. Afected areas
were irradiated with sunlight or an arc lamp. A total of
6 patients were treated, 4 of them achieved complete
resorption of lesions with no recurrence within 1 year
(1905). It was H. von Tappeiner (1904) who proposed
the term "photodynamic reaction" to describe a
specific photochemical reaction when irradiation with
light and the use of oxygen and a radiation-absorbing
dye, resulting in the death of biological systems [
15
].
Since the discovery of W.H. Hausmann new PS
hematoporphyrin (1908), the sensational experiment of
F. Meyer-Betz (1912) with a test of its efect on himself,
as well as after the works of A. Policard (1924), who
suggested the use of PDT in oncology, focusing on the
diagnostic value of fluorescence of tumor-transformed
tissues (using hematoporphyrin) [
14, 15
], the issue of
search and synthesis of new PS is relevant. Currently,
more than 100 drugs with photosensitizing activity have
been synthesized.
A new stage in the development of PDT began with
the medical use of lasers (1960s). Since then, PDT is
considered an intervention consisting in photochemical
destruction under combined exposure to PS and
low-intensity laser radiation with an optimal wavelength
corresponding to the absorption spectrum of the
sensitizer, which significantly increases the intensity of
photochemical reactions.
PDT has been developing as a direction of tumor
treatment for several decades. In one of the first reports
on PDT of glioma tissue (1980), it was assumed that
further improvement of the technique would promote
better penetration into tumor tissue and more radical
destruction of glioma cells [
16
]. The use of PDT in
neurooncology significantly increased the duration of overall
and relapse-free survival in patients, reduced the risk
of severe neurological complications [
17
]. In 1996, the
results of open phase I/II clinical trials involving more
than 310 patients who received PDT after resection of a
malignant tumor (primary or recurrent) were published.
A clear tendency to increase the average survival rate
of patients after surgical removal and single PDT was
revealed [
18, 19
].
In Ukraine, PDT as a new technology for the
treatment of oncological diseases began to be developed
in the 1970s [
20
]. Prof. M.F. Gamaliia and Prof.
R.E. Kavetsky substantiated the therapeutic effect
of laser irradiation by the presence of endogenous
porphyrins and molecular oxygen in the peripheral blood
as the main photoacceptors, formulating the concept
of "photodynamic blood modification" - optimized,
mathematically verified low-intensity efect on the blood
based on light parameters and doses, the consequences
of which are morphological changes of blood elements,
local and systemic efects. The developers were awarded
the Ukrainian SSR State Prize in the field of science and
technology [
21
].
The problem of using PDT in neuro-oncology
began to be addressed in the 1990s at the Institute
of Neurosurgery named after acad. A. P. Romodanov,
National Academy of Medical Sciences of Ukraine, under
the leadership of Prof. V.D. Rozumenko (neuro-oncology
department) with the involvement of the tissue culture
laboratory (head - Prof. V.M. Semenova). A number of
experimental studies [
22‒28
] formed the basis of the
clinical development of PDT regimens in neuro-oncology
patients [
29
]. Currently, PDT is a clinically acceptable
method of treating various types of cancer, which is
constantly being improved [
30, 31
].
Principle of photodynamic therapy and mechanism of action
Photodynamic therapy was introduced as an option
for topical (local) surgical therapy based on cytotoxic
efects caused by a chemical photosensitizing agent that
selectively accumulates in malignant tumor tissue and
is activated by light rays of the appropriate wavelength
[
32, 33
], generating singlet oxygen and free radicals,
which trigger photochemical reactions in tumor cells
with subsequent destruction of their protein structures.
Photodynamic therapy implies photoactivation of
PS molecule, which is selectively incorporated into
neoplastic cells [
34
].
Exposure to light rays of a certain wavelength
(photoirradiation) activates PS, exciting molecular
oxygen to the singlet or triplet state [
12
]. In the
singlet state, energy is converted into heat (internal
conversion) or emitted as light (fluorescence). Singlet
oxygen has a record long lifetime in the discharged state
— 4600 s, or 1.27 h [
36
]. In the triplet state, reactive
oxygen species (ROS) are generated, which react
rapidly with macromolecules containing unsaturated
double bonds (proteins, unsaturated fatty acids,
cholesterols), damaging the membranes of intracellular
organelles (mitochondria, lysosomes, endoplasmic
reticulum), reacting with DNA, proteins, lipids and
other macromolecules, disrupt multiple cell signaling
pathways, extensively destroy DNA, which eventually
leads to tumor cells death (through necrosis, apoptosis,
autophagy), destruction of the tumor microvascular
system, local ischemia, as well as to activation of
antitumor immune responses [
10, 12, 34, 37‒39
].
One of PDT targets is macrophages, which produce
inflammatory mediators and cytokines (lymphokines,
thromboxanes, prostaglandins, tumor necrosis factor,
etc.) after photoirradiation, thus significantly afecting
the tumor stroma degradation [
40
]. Moreover, PDT
damages endothelial cells, which leads to local
thrombosis, vasoconstric tion and ultimately to
the destruction of the microcirculatory channel.
The combination of these efects induces a strong
immune response against glioma in the experiment
[
41
]. Therefore, PDT engages the mechanisms of
not only direct cytodestructive, but also mediated
immunomodulatory antitumor efects, activating the
links of innate and acquired immunity.
Types of photosensitizers
Both PSs and photosensitizing precursors are used
for PDT [
10
]. Ideal PSs should be purified from impurities
in order to selectively accumulate in tumor tissue and be
able to cross the blood-brain barrier. Photosensitizers
should also be localized in the tumor tissue without
simultaneous accumulation in significant concentrations
in healthy tissue and have maximum cytotoxic activity
against tumor cells due to absorption of light photons
in the 650‒700 nm spectral range. It is important that
PSs do not cause systemic toxicity and are quickly
eliminated from the body [
42, 43
]. The selectivity of
PSs accumulation in the brain is one of the key issues
in the problem of PDT efectiveness increase. After
administration, PSs accumulate in all body organs, but
tumor tissue has a greater afinity for them [
35
]. The
blood-brain barrier, which prevents the penetration of
most drugs, does not interfere with PSs. The selectivity
of PSs accumulation in brain tissues varies from 3:1 to
50:1 compared to normal tissues [
42
].
Photosensitizers are divided into 1st, 2nd and 3rd
generation molecules.
The 1st generation PSs molecules are porphyrins,
namely por f imer sodium and hematoporphyrin
derivatives (HpD, a patented combination of monomers,
dimers and oligomers derived from hematoporphyrin)
[
10
]. In 1984, T.J. Dougherty et al. [
44
] conducted a
study on the isolation of the active fraction of HpD.
The purified mixture of HpD monomers, dimers and
oligomers was commercially named Photofrin (USA). In
Germany, the analogue is Photosun. These compounds
are characterized by strong absorption in the ~400 nm
range, but have limited excitation absorption at longer
light wavelengths [
45
]. HpD is an ineficient producer of
singlet oxygen, requiring prolonged photostimulation to
achieve an adequate therapeutic efect [
46
]. For PDT with
drugs based on HpD derivatives, laser radiation with a
wavelength of 628‒632 nm is used, while the depth of
photoinduced necrosis does not exceed 1 cm. Doses of
light energy vary significantly (from 50 to 500 J/cm²) and
depend on the size and localization of the tumor lesion.
The second generation photosensitizers such as
chlorins (thalaporfin sodium and temoporfin
(metatetra(hydroxyphenyl)chlorin (mTHPC)), pheophorbides,
bacteriopheophorbides, metalloporphyrins, purpurins,
phthalocyanines, and 5-aminolevulinic acid (5-ALA) have
been developed to overcome the limitations inherent to
1st generation sensitizers. They are usually activated
at wavelengths >600 nm and are more efective in
singlet oxygen production [
10
]. Thalaporfin sodium is
activated by light with a wavelength of 664 nm [
46, 47
].
Temoporfin (mTHPC) is the most potent of the clinically
available PSs (activated by 652 nm photostimulation),
is well tolerated, but has photosensitivity that persists
up to 6 weeks after administration. Another mTHPC
drug is Foscan (Germany). PDT requires minimal doses
(0.1–0.15 mg /kg of body weight) and light energy (10–20
J/cm²) at a wavelength of 652 nm. However, its use is
associated with side efects (development of tracheal
and bronchial stenosis, esophagotracheal fistulas,
esophageal perforations). The drug is rapidly excreted
from the body, skin toxicity is observed within 1 week
[
48, 49
]. Purlitin (selenium etiopurpurin) (USA) [
50
] and
Lutex (USA) belong to this group of PSs, the feature
of which is high selectivity of accumulation in tumors
[
51
]. Photosensitizers based on zinc and aluminum
porphyrazines and phthalocyanines are being studied
for their cytotoxic activity as new antitumor drugs [
52
].
5-ALA [
53
], which is physiologically an intermediate
product of heme synthesis, is used and commercially
available PSs of the 2nd generation. Clinical use is due to
oral bioavailability and a favorable safety profile. 5-ALA
is highly selective, mainly accumulating in the MG tissue
[
10
]. It is approved by the FDA for PDT of keratosis and
lfuorescence-guided visualization of tumor tissue during
surgical removal of MG [
10
]. Photodynamic therapy
based on 5-ALA is an efective method of adjuvant
treatment of MG. 5-ALA induces protoporphyrin IX
(PpIX) formation, what is rational to use during FGS
of MG due to the selective capture of photosensitizing
PpIX by the tumor and minimal skin sensitization [
54
].
However, insuficient accumulation of PpIX may limit the
possibility of using FGS and PDT in the marginal areas
of gliomas [
55
].
Photosensitizer s of the 3rd generation are
characterized by increased selectivity to tumor cells,
which is achieved due to conjugation with modifiers
(nanoparticles and antibodies) [
10, 46
]. When developing
the 3rd generation PSs, the focus was on creating
prodrugs that are activated only inside tumor cells. The
goal of the rational design of the 3rd generation PSs is
to reduce side efects while optimizing pharmacokinetics
and excitation absorption properties to maximize the
efective PDT window. New PSs from the groups of
polymethine and tricarbocyanine dyes, which have
some advantages over compounds of the porphyrin
series, are being studied. They are characterized by
a light absorption band with a high molar coeficient
in the spectrum range of 700‒900 nm. Currently, 3rd
generation PSs have not yet been approved for clinical
use for PDT in patients.
Experimental studies of photodynamic therapy
In vitro and in vivo experimental studies confirmed
the eficacy of PDT of brain tumors using 1st and 2nd
generation PSs (HpD, mTHPC, sinoporphyrin sodium, ZnPc
and Zn (II) phthalocyanines, tetraminephthalocyanine
(TAZnPc), 5-AL A, chlorin E6) either alone or in
combination with fractionated radiation therapy [
56
], cells), which matured, ac tivated and produced
sonodynamic therapy [
57
], temozolomide (TMZ) interleukin-6, which may be potentially used in cancer
chemotherapy (synergistic efect) [
58
] or EGFR inhibitors immunotherapy [
66
].
[
59, 60
]. In particular, the antitumor efect of PDT with PSs accumulation and PDT eficacy were evaluated
5-ALA in SU-DIPG-XIII MG cell cultures in combination in vitro in various glioma lines, primary neurons and
with fractionated radiation therapy was shown in vitro astrocytes of rats and in vivo in the healthy brain and
[
56
]. Phthalocyanines ZnPc and TAZnPc in vitro caused RG2 glioma of Fischer rats [
67
]. In vitro studies on
the death of GB cells of T98G, MO59, LN229 and rats showed a significant improvement in the survival
U87-MG lines by apoptosis, which was accompanied by of primary rat neuronal cells after PDT, in vivo studies
caspase-3 activation, loss of morphology and functions showed a decrease in the volume of edema/inaflmmation
of mitochondria, integrity of lysosomes, externalization on day 10 after PpIX-mediated PDT, confirming the
of phosphatidylserine and DNA fragmentation of tumor neuroprotective efect. Moderate hypothermia increased
cells [
61
]. Antitumor efects of combining PDT with PpIX fluorescence in the tumor by 5 times and the
sinoporphyrin sodium and sonodynamic therapy in average survival time of rats after PDT without afecting
vitro in U118 glioma cells were revealed [
57
]. Promising normal brain structures [
67
].
results were obtained for PDT with chlorin E6 of rat In an in vivo study, fox1 rnu/rnu rats with U87 GB
gliomas C6 and 101.8 in vivo [
62
]. after intraperitoneal injection of 5-ALA (100 mg/kg) were
Experimental study of PDT in vitro using 2nd interstitially irradiated to the tumor in two regimens:
generation PS - photosens (sulfonated aluminum with high (30 mW) or low (4.8 mW) radiation intensity.
phthalocyanine) was carried out at the Institute of Higher intensity and fractionation of irradiation caused
Neurosurgery in the tissue culture laboratory (Prof. a greater degree of tumor necrosis [
54
].
V.D. Rozumenko and Prof. V.M. Semenova). The method
of tissue culture was used to obtain the optimal efect Experimental approaches to enhancing the
of photodestruction of tumor cells in cultures of the efectiveness of photodynamic therapy
transplanted rat brain MG 101.8 and primary human Various preconditioning regimens are used to
MG cultures. Simultaneously, in a comparative aspect, enhance the fluorescence of PSs. Thus, selective
the reaction of cultured neural cells of the brain of enhancement of PpIX fluorescence and PDT with 5-ALA
experimental animals to the efect of PS and laser was achieved by pre-conditioning glioma cells and control
irradiation according to diferent schemes was studied. astrocytes with calcitriol for 48 h before incubation with
It was shown that laser irradiation with a wavelength 5-ALA. The authors suggest that the combined treatment
of 675 nm of cultures of experimental glioma 101.8 of glioma tumor cells with calcitriol using 5-ALA may be
pre-incubated in nutrient medium with the addition an efective and selective therapeutic method to improve
of photosens (5 μg/ml) causes photodestruction of the quality of PpIX fluorescence induced by 5-ALA [
55
].
most tumor cells, which rises with increasing exposure A combination of diferent treatment methods is
from 120 to 240‒300 s (subtotal tumor cell damage being investigated to increase the efectiveness of PDT.
and pronounced cell loss). Assessment of 24-h efect The synergistic efect of PDT and chemotherapy has
of PS on cultures of the normal brain of newborn rats been proved. It has been shown that the drug gefitinib,
demonstrated the resistance of neurocytes to PS EGFR inhibitor developed for treatment of breast and
accumulation and only weak incorporation of it into lung cancer, can enhance the photodynamic efect in
the cytoplasm of some glial cells, which confirms the brain tumor cells through gefitinib-mediated inhibition
absence of neurotoxicity when using it [
26, 27, 63
]. The of the ATP-binding cassette transporter ABCG2, which
results of these studies formed the basis of the clinical prevents the eflux of PS from brain tumor cells and
development of PDT regimens in neuro-oncological enhances efect of PDT [
59
]. The combined efect of
patients [
29, 64
]. another EGFR inhibitor, lapatinib, has been also studied.
In the study of the efect of PDT on gliosarcoma cells Its administration 24 h before PDT and for 3‒5 days
of the 9L/lacZ line in vitro using photodithazine (PDZ) and after PDT significantly increased PpIX accumulation
Biopdi/IRRAD-LED 660 LED light source at a wavelength and the decrease in LD50 after PpIX-mediated PDT in
of 660 nm showed 100% tumor cells death at diferent U87 and U87vIII MG cell lines in vitro and increased
concentrations of PS, which accumulated mainly in the the survival of U87 and GSC-30 glioma-grafted rats
area of nucleus and cytoplasm [
65
]. compared with therapy with lapatinib or PDT alone [
60
].
Photosens and PDZ at a light dose of 20 J/cm² The combination of TMZ with PDT significantly increased
(λex 615‒635 nm) efectively induced cell death of TMZ concentration in glioma tissues, enhanced tumor
glioma GL261 and fibrosarcoma MCA205 cells [
66
]. cell apoptosis, and increased the average survival rate
Photosens was predominantly localized in lysosomes, of rats with transplanted glioma [
58
].
and photoirradiation-induced cell death was inhibited One of the markers of tumor vessels in glioma is an
by apoptosis inhibitor (zVAD-fmk), ferostatin-1, and extra fibronectin domain A (ED A). The use of antibodies
ferroptosis inhibitors (DFO), but not by the necroptosis against ED A fibronectin promoted the accumulation
inhibitor necrostatin-1. Instead, PDZ accumulated of PS (F8-SIP) in the microvascular bed of the grafted
in the endoplasmic reticulum and Golgi apparatus, glioma SF126 in nude mice [
68
]. Subsequent PDT induced
and photochemical cell death was inhibited only microvascular stasis and thrombosis with decreased
by z-VAD-fmk. Tumor cells dying due to photosens functional density and decreased glioma growth with
or PDZ-induced photochemical reaction released restoration of microcirculation 4 days after treatment.
calreticulin, HMGB1 and ATP and were effectively Repeated use of PDT prevented microvascular recovery
absorbed by BMDCs (bone-marrow derived dendritic and resulted in a prolonged antiglioma efect [
68
].
The nature and role of ROS-mediated damage [
11
] involving 27 patients with newly diagnosed or
to tumor cells as a result of PDT have not been fully recurrent primary parenchymal brain tumors, among
elucidated. In this regard, the response of nitric oxide the 22 patients who were subsequently involved in the
(NO) signaling pathways in tumor cells is worth attention, study cohort, the 12-month total survival and 6-month
since the growth and proliferation of MG stem cells progression-free survival after surgery and PDT were
depend on the activation of inducible NO synthase (iNOS) 95.5 and 91.0%, respectively, in 13 patients with newly
[
69
], and iNOS inhibitors can play a role in increasing diagnosed GB - 100%. Side efects on the skin that can
PDT eficiency [
70
]. Thus, iNOS/NO of U87 glioblastoma be associated with the use of talaporfin sodium were
cells counteracts the cytotoxic efects of non-ionizing reported in 7.4% of patients. Skin photosensitivity test
PDT and stimulates the growth and migration activity results were relatively mild and completely disappeared
of surviving tumor cells [
71
], and iNOS-derived NO in within 15 days after administration of photosensitizer in
GB cells causes resistance to 5-ALA-based PDT [
72
]. all patients [
11
].
It is also known that tight intercellular junctions Two photosensitizing agents have been studied
(GJIC), formed by connexin (Cx) 43, can improve pr imar ily in MG: p or f imer s o dium (Photof r in,
"death signal" transmission between cells, increasing FDA-approved for the treatment of esophageal cancer)
the cytotoxic efect of chemotherapeutic agents, gene [
75
] and 5-ALA (FDA-approved for imaging of GB cells
therapy, and PDT [
73
]. Photodynamic therapy was more during surgery) [
13
]. In clinical trials, PDT was usually
efective in Cx43-transfected HeLa cells and Cx43- combined with other therapeutic interventions or
expressing U87 glioma cells at high cell density. This manipulations.
efect was significantly suppressed in the absence of Simultaneous use of fluorescence-guided surgery
Cx43 expression or Cx43-GJIC blockage. In addition, (FGS) and PDT allows both imaging of tumor cells and
in the presence of Cx43-GJIC, photofrin-PDT reduced selectively destruction of them [
34
]. For intraoperative
the mass of tumor xenografts in vivo. Increased PDT and postoperative PDT, special devices based on
fibereficiency associated with Cx43-GJIC was correlated with optic guided technique have been developed. Regardless
ROS-mediated stress signaling pathways, Ca2+ and lipid of PDT, PSs are used for auxiliary detection of the tumor
peroxidation [
73
]. Therefore, strategies to enhance Cx43 boundaries in order to maximize its removal during FGS
expression or Cx43-GJIC function may increase tumor (5-ALA, fluorescein, indocyanine green, endogenous
cell sensitivity to PDT and treatment eficacy. lfuorophores) [
76
].
One of the main caveats of PDT is that the laser
Clinical studies of photodynamic therapy beam must reach the cells containing the sensitizing
As mentioned above, PDT as a direction of tumor agent. Commonly used lasers have a wavelength range
treatment has been evolving for several decades. One of 630–690 nm, and the penetration depth rarely
of the first reports on PDT of glioma tissue in 1980 exceeds 5.0 mm in most tissues [
77
]. However, the
predicted that further improvement of the technique efects of PDT may be greater, as MRI data show the
would facilitate better penetration into tumor tissue and mean penetration depth of 9.1 mm [
78
] and postmortem
more radical destruction of glioma cells [
16
]. Although histopathological analysis revealed penetration up to
a large number of PDT trials have been conducted for 12.7 mm [
79
]. However, deeply located malignant cells
the treatment of malignant brain tumors, most of them remain completely unafected. In addition, according
are uncontrolled phase I or II studies, which difer to [
78
], 75% of cases of continued tumor growth/
greatly in their design (choice and dose of PS, choice recurrence were observed in the PDT area, which casts
and dose of photoirradiation, method of irradiation and doubt on the long-term efect even in the resection
use of additional techniques, choice and the formation of cavity. However, given the clinical availability of PSs and
groups of patients according to diagnoses, indicators and the low risk level of this therapy, further studies of their
interpretation of fndings, etc.), which makes the overall use in GB are needed [
80
].
analysis and assessment of treatment efectiveness A small number of clinical trials of PDT (porfimer
extremely dificult. sodium, 5-ALA) in the treatment of MG have been
In 1996, the results of open phase I and II clinical conducted, of which several have been discontinued or
trials, which involved more than 310 patients with withdrawn (n=3) or their status is unknown (n=3), 4 are
primary or recurrent malignant tumors, were published. ongoing [
81
].
Patients received PDT after tumor resection. A clear In a phase III trial using porfimer sodium for the
tendency was found to increase the average survival treatment of GB (n=27) [
82
], patients who received
rate of patients after surgical removal and single PDT ifve daily PDT sessions with an implanted laser in the
[
18
]. Based on the clinical experience of PDT for the resection cavity had better survival compared to those
treatment of gliomas in more than 350 patients using who did not receive PDT, but patients who received PDT
HpD (since 1987) and borated porphyrin (since 2001), a underwent resection with the help of FGS, which in itself
group of researchers from the Royal Melbourne Hospital improves treatment outcomes [
13
]. In addition, 15% of
reported an overall survival of patients with rediagnosed patients in both groups received TMZ, which limited the
and recurrent GB 28.0 and 40.0% after 2 years and 22.0 generalizability of the study results.
and 34.0% after 5 years. A meta-analysis of data from Another trial (phase I) used 5-ALA for FGS and
more than 1,000 patients with MG showed that median intraoperative PDT in 20 patients with recurrent GB
survival after PDT for rediagnosed and recurrent MG [
78
]. After 5-ALA FGS, 1‒4 cylindrical laser difusers
was 16.1 and 10.3 months, respectively [
74
]. According were inserted into the resection cavity and PDT was
to the results of a phase II clinical trial (JMA-IIA00026, performed under general anesthesia for 60 min (635
Japan) of intraoperative PDT with talaporfin sodium nm, difuser 200 mW/cm) with continuous irrigation
to maintain optical clarity and ventilation with 100% after the first relapse, 9 (20.5%) after the second,
oxygen. According to MRI performed after 24 hours, and another 9 (20.5%) after the third or subsequent
14 days and every 3 months thereafter, infection at the relapse. The mean iPDT target volume was 3.34 cm3
surgical site was noted in 1 patient was after 6 months (0.50–22.8 cm3). Severe neurological deterioration
as the only side efect. In 16 (80%) of 20 cases, cytotoxic lasted more than 6 weeks in only one patient. Median
edema along the resection margin was detected, which time-to-treatment-failure (TTF) was 7.1 months (95%
regressed or disappeared after 4–5 months. The median CI 4.4–9.8 months), median post-relapse survival
progression-free survival was 6 months (95% confidence 13.0 months (95% CI ‒ 9.2–16.8 months). The 2-year
interval (CI) – 4.8–7.2 months), which is comparable and 5-year post-relapse survival rates were 25.0 and
to the standard treatment for recurrent GB [
83
] and 4.5%, respectively. Response to treatment was not
confirms the innovativeness and safety of FGS and PDT significantly associated with patient characteristics,
combination with the use of 5 -ALA as a method of local treatment-related factors, or molecular markers. The
tumor control promising for tumor progression-free promising result and acceptable risk profile deserve
survival [
78
]. further prospective evaluation, particularly to determine
Preliminary results of a clinical study (phase I) the mechanisms and prognostic factors of a favorable
Intraoperative Photodynamic Therapy of GBM (INDYGO) response to the treatment of MG [
85
].
clinical trial (NCT03048240) involving 10 patients with In the study [
86
] the efectiveness of PDT with
primary GB, started in 2017 at the University Hospital talaporfin sodium in patients operated on for recurrent
of Lille (France), proved the safety and feasibility of GB was retrospectively evaluated. The average
relapseusing PDT based on 5-ALA in primary GB [
80
]. The free survival in patients with PDT (n=72) and the control
standardized treatment approach involved maximal group (n=38) after the second surgery was 5.7 and 2.2
resection (close to total or gross total tumor resection) months, respectively (p=0.0043), the average overall
under 5-ALA FGS control followed by intraoperative PDT. survival after the second surgery was 16, 0 and 12.8
In the postoperative period, patients received adjuvant months, respectively (p=0.031). In the PDT group, there
therapy (according to the Stupp protocol). Further was no significant diference in relapse-free and overall
follow-up included clinical examinations and brain MRI survival between patients previously diagnosed with GB
every 3 months until tumor progression and/or death. and those with malignant transformation to GB from less
No unacceptable or unexpected toxic or serious side malignant glioma variants. Therefore, the use of PDT
efects were reported. At the time of the interim analysis, in the progression of GB may have potential survival
the 12-month recurrence-free survival rate was 60% advantages, and its efectiveness is independent on
(median 17.1 months), the 12-month overall survival pre-recurrence pathology [
86
].
rate was 80% (median 23.1 months) [
80
]. Therefore, In another study [
87
] the clinical and surgical data
intraoperative administered PDT with 5-ALA immediately of the treatment of patients with primary GB, divided
after maximal resection as adjunctive therapy in primary into three groups were compared: operated on with the
GB is safe and may help reduce the risk of recurrence use of 5-ALA FGS followed by BCNU plates implantation
by targeting residual tumor cells in the resection cavity. (n=20, group I), those operated on with implantation
Other ongoing studies are using stereotactic PDT in of BCNU plates (n=42, group II), operated on with the
primary (NCT03897491) and recurrent (NCT04469699) use of 5-ALA FGS (n=59 patients, group III). In group
GB, as well as intraoperative and interstitial PDT (iPDT) I patients, life expectancy exceeded 3 years in 15%
in primary GB (NCT04391062, NCT03897491). of cases, the median relapse-free and overall survival
In the study [
84
], MRI results were analyzed in was 11 and 22 months, respectively. Patients implanted
11 patients with primary GB after iPDT. The procedure with BCNU plates had a significantly higher survival rate
implied the use of 5-ALA for selective metabolism of after tumor removal with 5-ALA FGS (22 months with
protoporphyrin IX (PpIX) in tumor cells and irradiation 5-ALA (Group I) vs. 18 months without 5-ALA (Group II),
using interstitially located optical cylindrical difuser p<0.0001). Patients in group I had an increased survival
ifbers (2‒10 fibers, difuser length 2-3 cm, 200 mW/cm, compared to group III (22 months with BCNU plates vs.
635 nm, 60 min irradiation), as well as intraoperative 21 months without BCNU plates, p=0.0025). Plate-related
spectral online monitoring. MRI results indicated adverse events were not significantly increased with
PDT-induced hemoglobin deoxygenation, methemoglobin 5-ALA FGS (20% with 5-ALA and 19% without 5-ALA) and
formation and were consistent with in vitro experiments. did not afect survival outcome. This study confirms the
The results of a retrospective study of the Medical synergistic efect in patients with primary GB of 5-ALA
Clinic of the University of Munich (Germany) on the FGS technology and BCNU plates implantation without
analysis of the risk profile of iPDT using induced 5-ALA increasing the incidence of adverse efects [
87
].
PpIX as a cytotoxic PS in a large monocentric cohort
of patients with local tumor recurrence after standard Therefore, PDT as a treatment method has a clearly
therapy (who were consecutively treated between 2006 focused efect aimed at selectively increasing the tumor
and 2018) proved its promise in local recurrences of destruction area during surgery, which improves the
MG [
85
]. 47 patients were treated, 44 of them were survival time and quality of life of patients with MG.
evaluated retrospectively. Recurrent gliomas included The main advantage of the PDT method is its high
37 GB (WHO grade 4) and 7 anaplastic astrocytomas eficiency, organ-sparing technique and low systemic
(WHO grade 3). Methylated O-6-methylguanine-DNA toxicity. In published studies of PDT efectiveness, there
methyltransferase (MGMT) was detected in 30 (68.2%) is no information on the development of resistance to
tumors, and wild-type isocitrate dehydrogenase (IDH) multiple PDT sessions, which suggests the possibility
in 29 (65.9%). 26 (59.1%) patients received treatment of re-treatment of tumor cells not removed during
surgery. The accumulated experience of PDT application
in oncological diseases suggests that PDT is one of
the most effective methods of stopping the local
spread of tumor cells, but it is necessary to optimize
radiation doses, the number of treatment procedures
and the intervals between them. The efectiveness of
photodynamic damage to a sensitized cell is determined
by the intracellular concentration of the sensitizer, its
localization in the cell, photochemical activity, and the
dose of laser irradiation. It is necessary to carry out
fundamental research with the development of new
experimental models of PDT, in which multiple efects
on the tumor will be applied.
Information disclosure
Conflict of interest
The authors declare no conflict of interest.
Ethical approval
This article is a literature review, therefore no ethics
committee approval was required.
Funding
The study was conducted without sponsorship.
1.
SungH
,
FerlayJ
,
SiegelRL
,
LaversanneM
,
SoerjomataramI
,
JemalA
,
BrayF
.
Global Cancer Statistics2020
:
GLOBOCAN Estimates of Incidence and Mortality Worldwide for 36 Cancers in 185 Countries
. CA
Cancer J Clin
.
2021
May;
71
(
3
):
209
-
249
. doi:
10
.3322/caac.21660
2.
LowJT
,
OstromQT
,
CiofiG
,
NefC
,
WaiteKA
,
KruchkoC
,
Barnholtz-SloanJS
.
Primary brain and other central nervous system tumors in the United States (2014
-2018)
: A summary of the CBTRUS statistical report for clinicians
.
Neurooncol Pract
.
2022 Feb22
;
9
(
3
):
165
-
182
. doi:
10
.1093/ nop/npac015
3.
LouisDN
,
PerryA
,
WesselingP
,
BratDJ
,
CreeIA
,
FigarellaBrangerD
,
HawkinsC
,
NgHK
,
PfisterSM
,
ReifenbergerG
,
SofiettiR
,
von DeimlingA
,
EllisonDW
.
The 2021 WHO Classification of Tumors of the Central Nervous System: a summary
.
Neuro Oncol. 2021; Aug2
;
23
(
8
):
1231
-
1251
. doi:
10
.1093/neuonc/noab106
4.
OstromQT
,
CiofiG
,
WaiteK
,
KruchkoC
,
Barnholtz-SloanJS
.
CBTRUS Statistical Report: Primary Brain and Other Central Nervous System Tumors Diagnosed in the United States in 2014-2018. Neuro Oncol. 2021 Oct5
;
23
(
12 Suppl 2
):
iii1
-
iii105
. doi:
10
.1093/neuonc/noab200
5.
FedorenkoZ
,
MichailovichYu
,
GoulakL
,
GorokhYe
,
RyzhovA
,
SoumkinaO
,
Koutsenko L. CANCER INUKRAINE
,
2020
-
2021
:
Incidence, mortality, prevalence and other relevant statistics
.
Bulletin of the National Cancer Registry of Ukraine
.
2022
;23. Available at: http://www.ncru.inf.ua/ publications/BULL_23/index_e.htm
6.
KliuchkaVM
,
RozumenkoAV
,
RozumenkoVD
,
SemenovaVM
,
MalyshevaTA
. [
Heterogeneity of oligoastrocytoma: morphology, surgery, and survival in the series of 163 patients
. Retrospective study].
Ukrainian Neurosurgical Journal
.
2018
;(3):
24
-
33
. Ukrainian. doi:
10
.25305/ unj.126654
7.
RozumenkoAV
,
KliuchkaVM
,
RozumenkoVD
,
FedorenkoZP
. [
Survival rates in patients with the newly diagnosed glioblastoma: Data from the National Cancer Registry of Ukraine,2008
-
2016
].
Ukrainian Neurosurgical Journal
.
2018
;(2):
33
-
9
. Ukrainian. doi:
10
.25305/unj.124878
8.
DupontC
,
VermandelM
,
LeroyHA
,
QuidetM
,
LecomteF
,
DelhemN
,
MordonS
,
ReynsN.Intraoperative photoDynamic Therapy for Glioblastomas (INDYGO): Study Protocol for a Phase I Clinical Trial
.
Neurosurgery. 2019 Jun1
;
84
(
6
):
E414
-
E419
. doi:
10
.1093/neuros/nyy324
9.
van SolingeTS
,
NielandL
,
ChioccaEA
,
BroekmanMLD
.
Advances in local therapy for glioblastoma - taking the fight to the tumour
.
Nat Rev Neurol
.
2022
; Apr;
18
(
4
):
221
-
236
. doi:
10
.1038/s41582-022-00621-0
10.
MahmoudiK
,
GarveyKL
,
BourasA
,
CramerG
,
SteppH
,
Jesu RajJG
,
BozecD
,
BuschTM
,
HadjipanayisCG
. 5
-aminolevulinic acid photodynamic therapy for the treatment of high-grade gliomas
.
J Neurooncol
.
2019
Feb;
141
(
3
):
595
-
607
. doi:
10
.1007/s11060-019-03103-4
11.
MuragakiY
,
AkimotoJ
,
MaruyamaT
,
IsekiH
,
IkutaS
,
NittaM
,
MaebayashiK
,
SaitoT
,
OkadaY
,
KanekoS
,
MatsumuraA
,
KuroiwaT
,
KarasawaK
,
NakazatoY
,
KayamaT. PhaseII clinical study on intraoperative photodynamic therapy with talaporfin sodium and semiconductor laser in patients with malignant brain tumors
.
J Neurosurg
.
2013
Oct;
119
(
4
):
845
-
52
. doi:
10
.3171/
2013
.7.JNS13415
12.
QuirkBJ
,
BrandalG
,
DonlonS
,
VeraJC
,
MangTS
,
FoyAB
,
LewSM
,
GirottiAW
,
JogalS
,
LaViolettePS
,
ConnellyJM
,
Whelan HT
.
Photodynamic therapy (PDT) for malignant brain tumors--where do we stand? Photodiagnosis Photodyn Ther
.
2015
Sep;
12
(
3
):
530
-
44
. doi:
10
.1016/j. pdpdt.
2015
.
04
.009
13.
StummerW
,
PichlmeierU
,
MeinelT
,
WiestlerOD
,
ZanellaF
,
ReulenHJ
;
ALA-GliomaStudyGroup
.
Fluorescenceguided surgery with 5-aminolevulinic acid for resection of malignant glioma: a randomised controlled multicentre phase III trial
.
Lancet Oncol
.
2006
; May;
7
(
5
):
392
-
401
. doi:
10
.1016/S1470-2045(
06
)
70665
-
9
14.
WyssP
,
TadirY
,
TrombergBJ
,
HallerU
.
History of photomedicine
.
Photomedicine in Gynecology and Reproduction
. Basel: Karger,
2000
. p.
4
-
11
.
15.
NazarianRS
,
SpіrіdonovaKYu
,
PіontkovskaOV
,
VlasovAV
.
[Photodynamic Therapy: From the Antiquity to the Present
. Literature Review].
Novini stomatologії
.
2015
;(3):
68
-
70
. Ukrainian.
16.
PerriaC
,
CapuzzoT
,
CavagnaroG
,
DattiR
,
FrancavigliaN
,
RivanoC
,
TerceroVE
.
Fast attempts at the photodynamic treatment of human gliomas
.
J Neurosurg Sci
.
1980
JulDec;
24
(
3-4
):
119
-
29
17.
MullerPJ
,
WilsonBC
.
Photodynamic therapy for malignant newly diagnosed supratentorial gliomas
.
J Clin Laser Med Surg
. 1996 Oct;
14
(
5
):
263
-
70
. doi:
10
.1089/clm.
1996
.
14
.263
18.
KostronH
,
ObwegeserA
,
JakoberR.Photodynamic therapy in neurosurgery: a review
.
J Photochem Photobiol B
.
1996
Nov;
36
(
2
):
157
-
68
. doi:
10
.1016/s1011-
1344
(
96
)
07364
-
2
19.
KostronH
.
Photodynamic diagnosis and therapy and the brain
.
Methods Mol Biol
.
2010
;
635
:
261
-
80
. doi:
10
.1007/978-1-
60761
-697-9_
17
20.
GamaleyaNF.
[
Lasers in experiment
and clinic]. Moscow: Medicine;
1972
. Russian.
21.
Zavads'kaTS
,
BoykoII
,
BoykoAH
.
Fotodynamichna terapiya v onkolohiyi
.
Oncology
.
2021
;(
1-2
):
53
-
56
. Ukrainian.
22.
BogachkovNI
,
MelnikIS
,
RozumenkoVD
.
Tekhnika i metodologiya fotodinamicheskoy terapii
.
T.1
. Lazery v terapii zlokachestvennykh opukholey: Kiev: Kievskiy politekhnicheskiy institute;
1995
. Russian.
23.
BidnenkoVN
,
SigalVL
,
RozumenkoVD
.
Efekty lokal'noy gipertermii pri fotodinamicheskoy terapii opukholey mozga
.
Dopovidi NAN Ukrayiny
.
1999
;(10):
181
-
85
. Russian.
24.
BidnenkoVN
,
SigalVL
,
RozumenkoVD
.
Theoretical estimations of the area of destruction in brain tumors under photodynamic therapy
.
Proc. SPIE 4162
,
Controlling Tissue Optical Properties: Applications in Clinical Study, (3 Nov.2000
); doi: 10.1117/12.405938.
25.
NosovAT
,
RozumenkoVD
,
SemenovaVM
,
MedyanikІO
.
Morfofunktsіonal'nі zmіni mozku pri dії vipromіnyuvannya visokoenergetichnikh vuglekislotnogo, neodimovogo-AІG ta gol'mієvogo lazerіv
.
Byulleten' UAN
.
1998
;(5):
136
-
137
. Ukrainian.
26.
RozumenkoV.D.
,
SemenovaV.M.Fotodinamicheskaya terapiya opukholey golovnogo mozga: efekt v kul'ture gliomy (shtamm 101.8) s primeneniem ftalotsianina
.
Fotobiolohiya i fotomedytsyna
.
2000
;
1
(
2
):
65
-
70
. Russian.
27.
RozumenkoVD
,
SemyonovaVM
,
StajnoLP
,
GerasenkoKM
.
Photodynamic therapy of brain tumours:efect in culture of glioma (stamm 101.8
).
Ukrainian Neurosurgical Journal
.
2001
;(4):
59
-
66
. Russian.
28.
RozumenkoVD
,
SemenovaVM
,
OthmanO.
[
The brain and glial tumors tissue morphology changes under the highly energetic radiation of CO2 and Nd-YAG lasers influence]
.
Ukrainian Neurosurgical Journal
.
2004
;(3):
36
-
42
. Russian.
29.
RozumenkoVD
,
SihalVL
,
KhomenkoOV
,
HerasenkoKM
.
Sposib terapiyi hlybokoroztashovanykh pukhlynnykh tkanyn
.
Patent of Ukraine 49273
.
2002
September 16. Ukraine.
30.
B e r e z o v s ' k a I V, B i l a s h O M , R o z h y t s ' k y y M M . Fotosensibilizator dlya fotodynamichnoyi terapiyi
.
Patent of Ukraine 87864
.
2014
February 25. Ukraine.
31.
KholinVV
.
Sposib oprominennya pukhlyny metodom lazernoho skanuvannya v fotodynamichniy terapiyi
.
Patent of Ukraine 89226
.
2014
April 10. Ukraine.
32.
BeckTJ
,
KrethFW
,
BeyerW
,
MehrkensJH
,
ObermeierA
,
SteppH
,
StummerW
,
BaumgartnerR
.
Interstitial photodynamic therapy of nonresectable malignant glioma recurrences using 5-aminolevulinic acid induced protoporphyrin IX
.
Lasers Surg Med
.
2007
; Jun;
39
(
5
):
386
-
93
. doi:
10
.1002/lsm.20507
33.
DubeySK
,
PradyuthSK
,
SahaRN
,
SinghviG
,
AlexanderA
,
AgrawalM
,
ShapiroBA
,
PuriA
.
Application of photodynamic therapy drugs for management of glioma
.
Journal of Porphyrins and Phthalocyanines
.
2019
;
23
(
11
-12):
1216
-
28
. doi:
10
.1142/S1088424619300192.
34.
CramerSW
,
ChenCC
.
Photodynamic Therapy for the Treatment of Glioblastoma
.
Front Surg
.
2020 Jan21
;6:
81
. doi:
10
.3389/fsurg.
2019
.00081
35.
GelfondML
,
BarchukAS
,
VasilievDT
,
StukovAN
.
[PDT opportunities in oncology practice]
.
Russian Journal of Biotherapy
.
2003
;
2
(
4
):
67
-
71
. Russian.
36.
MinaevBF.
[
Spin-catalysis in the processes of photo- and bioactivation of molecular oxygen]
.
Ukr Biokhim Zh
(
1999
). 2009 May-Jun;
81
(
3
):
21
-
45
. Russian.
37.
CastanoAP
,
DemidovaTN
,
HamblinMR
.
Mechanisms in photodynamic therapy: part two- cellular signaling, cell metabolism and modes of cell death
.
Photodiagnosis Photodyn. Ther
.
2005
;(2):
1
-
23
. doi:
10
.1016/S1572-
1000
(
05
)
00030
-X
38.
AgostinisP
,
BergK
,
CengelKA
,
FosterTH
,
GirottiAW
,
GollnickSO
,
HahnSM
,
HamblinMR
,
JuzenieneA
,
KesselD
,
KorbelikM
,
MoanJ
,
MrozP
,
NowisD
,
PietteJ
,
WilsonBC
,
GolabJ.Photodynamic therapy of cancer: an update
.
CA Cancer J Clin
.
2011
Jul-Aug;
61
(
4
):
250
-
81
. doi:
10
.3322/ caac.20114
39.
KanekoS
,
FujimotoS
,
YamaguchiH
,
YamauchiT
,
YoshimotoT
,
TokudaK.Photodynamic Therapy of Malignant Gliomas
.
Prog Neurol Surg
.
2018
;
32
:
1
-
13
. doi:
10
.1159/000469675
40.
MarksPV
,
IgbaseimokumoU
,
Chakrabarty А
.
A preliminary experimental in vivo study of the efect of photodynamic therapy on human pituit ar y adenoma implanted in mice
.
Br. J. Neurosurg
.
1998
;
12
(
2
):
140
-
5
. doi:
10
.1080/02688699845285
41.
LiF
,
ChengY
,
LuJ
,
HuR
,
WanQ
,
FengH.Photodynamic therapy boosts anti-glioma immunity in mice: a dependence on the activities of T cells and complement C3
.
J Cell Biochem
.
2011
; Oct;
112
(
10
):
3035
-
43
. doi:
10
.1002/ jcb.23228
42.
MullerPJ
,
WilsonBC
.
Photodynamic therapy of brain tumors--a work in progress
.
Lasers Surg Med
. 2006 Jun;
38
(
5
):
384
-
9
. doi:
10
.1002/lsm.20338. PMID:
16788926
.
43.
ZavadskayaТS
.
Photodynamic therapy in the treatment of glioma
.
Exp Oncol
.
2015
; Dec;
37
(
4
):
234
-
41
44.
DoughertyTJ
,
PotterWR
,
WeishauptKR
.
The structure of the active component of hematoporphyrin derivative
. In: Dorion D.R., Gomer C.J., editors.
Porphyrin localization and treatment of tumors
. New York:
Alan R Liss Inc
.
1984
:
301
-
314
. Available at: https://www.pdt-association.com/history.
45.
de PaulaLB
,
PrimoFL
,
TedescoAC
.
Nanomedicine associated with photodynamic therapy for glioblastoma treatment
.
Biophys Rev
.
2017
Oct;
9
(
5
):
761
-
773
. doi:
10
.1007/s12551-017-0293-3
46.
AllisonRR
,
SibataCH
.
Oncologic photodynamic therapy photosensitizers: a clinical review
.
Photodiagnosis Photodyn Ther
.
2010 Jun;7
(
2
):
61
-
75
. doi:
10
.1016/j. pdpdt.
2010
.
02
.001
47.
WangS
,
BromleyE
,
XuL
,
ChenJC
,
KeltnerL
.
Talaporfin sodium
.
Expert Opin Pharmacother
.
2010
Jan;
11
(
1
):
133
-
40
. doi:
10
.1517/14656560903463893
48.
MüllerS
,
WaltH
,
Dobler-GirdziunaiteD
,
FiedlerD
,
HallerU
.
Enhanced photodynamic efects using fractionated laser light
.
J Photochem Photobiol B
.
1998
Jan;
42
(
1
):
67
-
70
. doi:
10
.1016/S1011-
1344
(
97
)
00124
-
3
49.
VeenhuizenRB
,
RuevekampMC
,
OppelaarH
,
HelmerhorstTJ
,
KenemansP
,
StewartFA
.
Foscan-mediated photodynamic therapy for a peritoneal-cancer model: drug distribution and eficacy studies
.
Int J Cancer
.
1997 Oct9
;
73
(
2
):
230
-
5
. doi:
10
.1002/(sici)
1097
-
0215
(
19971009
)73:
2
<
230
::aidijc12>
3
.0.co;2-
j
50.
KaplanMJ
,
Somer sRG
,
GreenbergRH
,
AcklerJ.Photodynamic therapy in the management of metastatic cutaneous adenocarcinomas: case reports from phase 1/2 studies using tin ethyl etiopurpurin (SnET2)
.
J Surg Oncol
.
1998
Feb;
67
(
2
):
121
-
5
. doi:
10
.1002/(sici)
1096
-
9098
(
199802
)67:
2
<
121
.
:
aid-jso9>
3
.0.
c
51.
RenschlerM
,
YuenA
,
PanellaT.Photodynamic therapy trials with Lutetium Texaphyrin
.
Photochem Photobiol
.
1997
;
65
:
475
.
52.
MachacekM
,
KollarJ
,
MiletinM
,
KuceraR
,
KubatP
,
SimunekT
,
NovakovaV
,
ZimcikP
.
Anionic hexadeca-carboxylate tetrapyrazinoporphyrazine: synthesis and in vitro photodynamic studies of water-soluble non-aggregating photosensitizer
.
RSC Advances
.
2016
;
6
(
12
):
10064
-
77
. doi 10.1039/C5RA25881B
53.
SteppH
, Stummer W. 5
-ALA in the management of malignant glioma
.
Lasers Surg Med
. 2018 Jul;
50
(
5
):
399
-
419
. doi:
10
.1002/lsm.22933
54.
TetardMC
,
VermandelM
,
MordonS
,
LejeuneJP
,
ReynsN.Experimental use of photodynamic therapy in high grade gliomas: a review focused on 5-aminolevulinic acid
.
Photodiagnosis Photodyn Ther
.
2014
Sep;
11
(
3
):
319
-
30
. doi:
10
.1016/j.pdpdt.
2014
.
04
.004
55.
ChenX
,
WangC
,
TengL
,
LiuY
,
ChenX
,
YangG
,
WangL
,
LiuH
,
LiuZ
,
ZhangD
,
ZhangY
,
GuanH
,
LiX
,
FuC
,
ZhaoB
,
YinF
,
ZhaoS
.
Calcitriol enhances 5-aminolevulinic acid-induced lfuorescence and the efect of photodynamic therapy in human glioma
.
Acta Oncol
.
2014
Mar;
53
(
3
):
405
-
13
. doi:
10
.3109/0284186X.
2013
.819993
56.
SkandalakisGS
,
BourasA
,
RiveraD
,
RizeaC
,
RajJG
,
BozecD
,
HadjipanayisCG
.
Photodynamic Therapy of Difuse Intrinsic Pontine Glioma in Combination with Radiation
.
Neurosurgery
. 2020 Dec;
67
(
Supplement
_1):nyaa447_
873
. doi:
10
.1093/neuros/nyaa447_
873
57.
AnYW
,
LiuHQ
,
ZhouZQ
,
WangJC
,
JiangGY
,
LiZW
,
WangF
,
JinHT
.
Sinoporphyrin sodium is a promising sensitizer for photodynamic and sonodynamic therapy in glioma
.
Oncol Rep
. 2020 Oct;
44
(
4
):
1596
-
1604
. doi:
10
.3892/or.
2020
.7695
58.
ZhangX
,
GuoM
,
ShenL
,
HuS.Combination of photodynamic therapy and temozolomide on glioma in a rat C6 glioma model
.
Photodiagnosis Photodyn Ther
.
2014
Dec;
11
(
4
):
603
-
12
. doi:
10
.1016/j.pdpdt.
2014
.
10
.007
59.
SunW
,
KajimotoY
,
InoueH
,
MiyatakeS
,
IshikawaT
,
KuroiwaT.Gefitinib enhances the eficacy of photodynamic therapy using 5-aminolevulinic acid in malignant brain tumor cells
.
Photodiagnosis Photodyn Ther
.
2013
Feb;
10
(
1
):
42
-
50
. doi:
10
.1016/j.pdpdt.
2012
.
06
.003
60.
FisherC
,
ObaidG
,
NiuC
,
FoltzW
,
GoldsteinA
,
HasanT
,
LilgeL. LiposomalLapatinib in Combination with LowDose Photodynamic Therapy for the Treatment of Glioma
.
Journal of Clinical Medicine
.
2019
;
8
(
12
):
2214
. doi:
10
.3390/ jcm8122214
61.
VelazquezFN
,
MirettiM
,
BaumgartnerMT
,
CaputtoBL
,
TempestiTC
,
PruccaCG
.
Efectiveness of ZnPc and of an amine derivative to inactivate Glioblastoma cells by Photodynamic Therapy: an in vitro comparative study
.
Sci Rep
.
2019
;
9
:3010 doi: 10.1038/s41598-019-39390-0
62.
ZavadskayaTS
,
TaranetsLP
,
TrompakOO
. [
Fotolonmediated photodynamic therapy of experimental gliomas]
.
Photobiology and Photomedicine
.
2013
;(
1-2
):
81
-
8
. Ukrainian. Available at: http://fnfjournal.univer.kharkov. ua/Ru/nomera/2013_
1_2/zavadska
.pdf
63.
RozumenkoV.D.
,
SemenovaV.M.
,
StaynoL.P.Issledovanie effekta fotodinamicheskoy terapii v kul'turakh gliom golovnogo mozga eksperimental'nykh zhivotnykh i cheloveka. Aspekty primeneniya metoda kul'tivirovaniya tkaney v neyrobiologii i neyroonkologii
. Kiev: Interservis;
2018
. Russian.
64.
RozumenkoVD
.
Innovative laser technologies in brain tumors surger y
.
Photobiology аnd Photomedicine
.
2018
;
24
:
9
-
12
. Available at: http://fnfjournal.univer. kharkov.ua/Ua/nomera/2018_1/1_rozumenko.pdf
65.
FontanaLC
,
PintoJG
,
PereiraAHC
,
SoaresCP
,
RanieroLJ
,
Ferreira-StrixinoJ
.
Photodithazine photodynamic efect on viability of 9L/lacZ gliosarcoma cell line
.
Lasers Med Sci
.
2017
Aug;
32
(
6
):
1245
-
1252
. doi:
10
.1007/s10103-017- 2227-5
66.
TurubanovaVD
,
BalalaevaIV
,
MishchenkoTA
,
CatanzaroE
,
AlzeibakR
,
PeskovaNN
,
EfimovaI
,
BachertC
,
MitroshinaEV
,
KryskoO
,
VedunovaMV
,
KryskoDV
.
Immunogenic cell death induced by a new photodynamic therapy based on photosens and photodithazine
.
J Immunother Cancer
.
2019 Dec16
;
7
(
1
):
350
. doi:
10
.1186/s40425-019-0826-3
67.
FisherCJ
,
NiuC
,
FoltzW
,
ChenY
,
Sidorova-DarmosE
,
EubanksJH
,
LilgeL
.
ALA-PpIX mediated photodynamic therapy of malignant gliomas augmented by hypothermia
.
PLoS One
.
2017 Jul31
;
12
(
7
):e0181654. doi:
10
.1371/ journal.pone.0181654
68.
AckerG
,
PalumboA
,
NeriD
,
VajkoczyP
,
CzabankaM.F8-SIP mediated targeted photodynamic therapy leads to microvascular dysfunction and reduced glioma growth
.
J Neurooncol
.
2016
Aug;
129
(
1
):
33
-
8
. doi:
10
.1007/s11060- 016-2143-8
69.
EylerCE
,
WuQ
,
YanK
,
MacSwordsJM
,
Chandler-MilitelloD
,
MisuracaKL
,
LathiaJD
,
ForresterMT
,
LeeJ
,
StamlerJS
,
GoldmanSA
,
BredelM
,
McLendonRE
,
SloanAE
,
HjelmelandAB
,
RichJN
.
Glioma stem cell proliferation and tumor growth are promoted by nitric oxide synthase-2
. Cell.
2011 Jul8
;
146
(
1
):
53
-
66
. doi:
10
.1016/j.cell.
2011
.
06
.006
70.
BhowmickR
,
GirottiAW
.
Pro-survival and pro-growth efects of stress-induced nitric oxide in a prostate cancer photodynamic therapy model
.
Cancer Lett
.
2014 Feb1
;
343
(
1
):
115
-
22
. doi:
10
.1016/j.canlet.
2013
.
09
.025
71.
FaheyJM
,
KorytowskiW
,
GirottiAW
.
Upstream signaling events leading to elevated production of pro-survival nitric oxide in photodynamically-challenged glioblastoma cells
.
Free Radic Biol Med
. 2019 Jun;
137
:
37
-
45
. doi:
10
.1016/j. freeradbiomed.
2019
.
04
.013
72.
FaheyJM
,
Girotti AW
.
Nitric Oxide Antagonism to AntiGlioblastoma Photodynamic Therapy: Mitigation by Inhibitors of Nitric Oxide Generation
.
Cancers (Basel)
.
2019 Feb15
;
11
(
2
):
231
. doi:
10
.3390/cancers11020231
73.
WuDP
,
BaiLR
,
LvYF
,
ZhouY
,
DingCH
,
YangSM
,
ZhangF
,
Huang JL
.
A novel role of Cx43-composed GJIC in PDT phototoxicity: an implication of Cx43 for the enhancement of PDT eficacy
.
Int J Biol Sci. 2019 Jan1
;
15
(
3
):
598
-
609
. doi:
10
.7150/ijbs.29582
74.
ChernovMF
,
MuragakiY
,
KesariS
,
McCutcheonIE
(eds): Intracranial Gliomas.
Part III - Innovative Treatment Modalities. Prog Neurol Surg
. Basel, Karger.
2018
;
32
:
1
-
13
. doi:
10
.1159/000469675.
75.
BellnierDA
,
GrecoWR
,
LoewenGM
,
NavaH
,
OserofAR
,
DoughertyTJ.Clinical pharmacokinetics of the PDT photosensitizers porfimer sodium (Photofrin
),
2
-[1-hexyloxyethyl]
-2-devinyl pyropheophorbide-a (Photochlor) and 5-AL A-induced protoporphyrin IX
.
Lasers Surg Med
.
2006
; Jun;
38
(
5
):
439
-
44
. doi:
10
.1002/ lsm.20340. PMID:
16634075
.
76.
SendersJT
,
MuskensIS
,
SchnoorR
,
KarhadeAV
,
CoteDJ
,
SmithTR
,
BroekmanML
.
Agents for fluorescence-guided glioma surgery: a systematic review of preclinical and clinical results
.
Acta Neurochir (Wien)
.
2017
Jan;
159
(
1
):
151
-
167
. doi:
10
.1007/s00701-016-3028-5
77.
WangHW
,
ZhuTC
,
PuttME
,
SolonenkoM
,
MetzJ
,
DimofteA
,
MilesJ
,
FrakerDL
,
GlatsteinE
,
HahnSM
,
YodhAG
.
Broadband reflectance measurements of light penetration, blood oxygenation, hemoglobin concentration, and drug concentration in human intraperitoneal tissues before and after photodynamic therapy
.
J Biomed Opt
.
2005
; JanFeb;
10
(
1
):
14004
. doi:
10
.1117/1.1854679
78.
SchipmannS
,
MütherM
,
StögbauerL
,
ZimmerS
,
BrokinkelB
,
HollingM
,
GrauerO
,
Suero MolinaE
,
WarnekeN
,
StummerW.Combination of ALA-induced fluorescence-guided resection and intraoperative open photodynamic therapy for recurrent glioblastoma: case series on a promising dual strategy for local tumor control
.
J Neurosurg
.
2020; Jan24
:
1
-
11
. doi:
10
.3171/
2019
.11.JNS192443
79.
AkimotoJ
,
FukamiS
,
SudaT
,
IchikawaM
,
HaraokaR
,
KohnoM
,
Shishido-HaraY
,
NagaoT
,
KurodaM.First autopsy analysis of the eficacy of intra-operative additional photodynamic therapy for patients with glioblastoma
.
Brain Tumor Pathol
.
2019
; Oct;
36
(
4
):
144
-
151
. doi:
10
.1007/ s10014-019-00351-0
80.
VermandelM
,
DupontC
,
LecomteF
,
LeroyHA
,
TuleascaC
,
MordonS
,
HadjipanayisCG
,
ReynsN.Standardized intraoperative 5-ALA photodynamic therapy for newly diagnosed glioblastoma patients: a preliminary analysis of the INDYGO clinical trial
.
J Neurooncol
.
2021
; May;
152
(
3
):
501
-
514
. doi:
10
.1007/s11060-021-03718-6
81. Search of: photodynamic therapy | Glioma - List
Results
[Internet].
U.S. National Library of Medicine. ClinicalTrials
. gov. Available at: https://clinicaltrials.gov/ct2/results?con d=
Glioma&term=photodynamic++therapy&cntry=&state =&city=&dist=&Search=Search
82.
EljamelMS
,
GoodmanC
,
MoseleyH. ALAand Photofrin fluorescence-guided resection and repetitive PDT in glioblastoma multiforme: a single centre Phase III randomised controlled trial
.
Lasers Med Sci
.
2008
Oct;
23
(
4
):
361
-
7
. doi:
10
.1007/s10103-007-0494-2
83.
van LindeME
,
BrahmCG
,
de Witt HamerPC
,
ReijneveldJC
,
BruynzeelAME
,
VandertopWP
,
van de VenPM
,
WagemakersM
,
van der WeideHL
,
EntingRH
,
WalenkampAME
,
VerheulHMW
.
Treatment outcome of patients with recurrent glioblastoma multiforme: a retrospective multicenter analysis
.
J Neurooncol
.
2017
; Oct;
135
(
1
):
183
-
192
. doi:
10
.1007/s11060-017-2564-z
84.
AumillerM
,
HecklC
,
QuachS
,
SteppH
,
Ertl-WagnerB
,
SrokaR
,
ThonN
,
RühmA
.
Interrelation between Spectral Online Monitoring and Postoperative T1-Weighted MRI in Interstitial Photodynamic Therapy of Malignant Gliomas
.
Cancers (Basel)
.
2021; Dec27
;
14
(
1
):
120
. doi:
10
.3390/ cancers14010120
85.
LietkeS
,
SchmutzerM
,
SchwartzC
,
WellerJ
,
SillerS
,
AumillerM
,
HecklC
,
ForbrigR
,
NiyaziM
,
EgenspergerR
,
SteppH
,
SrokaR
,
TonnJC
,
RühmA
,
ThonN.Interstitial Photodynamic Therapy Using 5-ALA for Malignant Glioma Recurrences
.
Cancers (Basel)
.
2021; Apr7
;
13
(
8
):
1767
. doi:
10
.3390/cancers13081767
86.
KobayashiT
,
NittaM
,
ShimizuK
,
SaitoT
,
TsuzukiS
,
FukuiA
,
KoriyamaS
,
KuwanoA
,
KomoriT
,
MasuiK
,
MaeharaT
,
KawamataT
,
MuragakiY.TherapeuticOptions for Recurrent Glioblastoma-Eficacy of Talaporfin Sodium Mediated Photodynamic Therapy
.
Pharmaceutics. 2022; Feb2
;
14
(
2
):
353
. doi:
10
.3390/pharmaceutics14020353
87.
Della PuppaA
,
LombardiG
,
RossettoM
,
RustemiO
,
BertiF
,
CecchinD
,
GardimanMP
,
RolmaG
,
PersanoL
,
ZagonelV
,
ScienzaR
.
Outcome of patients afected by newly diagnosed glioblastoma undergoing surgery assisted by 5-aminolevulinic acid guided resection followed by BCNU wafers implantation: a 3-year follow-up
.
J Neurooncol
.
2017
; Jan;
131
(
2
):
331
-
340
. doi:
10
.1007/s11060-016-2301-z