Next Article in Journal
Chlorotoxin and Lung Cancer: A Targeting Perspective for Drug Delivery
Next Article in Special Issue
CDR3 Variants of the TXB2 Shuttle with Increased TfR1 Association Rate and Enhanced Brain Penetration
Previous Article in Journal
Folate-Functionalization Enhances Cytotoxicity of Multivalent DNA Nanocages on Triple-Negative Breast Cancer Cells
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Photodynamic Opening of the Blood–Brain Barrier and the Meningeal Lymphatic System: The New Niche in Immunotherapy for Brain Tumors

by
Oxana Semyachkina-Glushkovskaya
1,2,*,
Andrey Terskov
1,
Alexander Khorovodov
1,
Valeria Telnova
1,
Inna Blokhina
1,
Elena Saranceva
1 and
Jürgen Kurths
1,3
1
Department of Biology, Saratov State University, Astrakhanskaya Str. 83, 410012 Saratov, Russia
2
Institute of Physics, Humboldt University, Newtonstrasse 15, 12489 Berlin, Germany
3
Department of Complexity Science, Potsdam Institute for Climate Impact Research, Telegrafenberg A31, 14473 Potsdam, Germany
*
Author to whom correspondence should be addressed.
Pharmaceutics 2022, 14(12), 2612; https://doi.org/10.3390/pharmaceutics14122612
Submission received: 7 October 2022 / Revised: 13 November 2022 / Accepted: 24 November 2022 / Published: 26 November 2022

Abstract

:
Photodynamic therapy (PDT) is a promising add-on therapy to the current standard of care for patients with glioblastoma (GBM). The traditional explanation of the anti-cancer PDT effects involves the PDT-induced generation of a singlet oxygen in the GBM cells, which causes tumor cell death and microvasculature collapse. Recently, new vascular mechanisms of PDT associated with opening of the blood–brain barrier (OBBB) and the activation of functions of the meningeal lymphatic vessels have been discovered. In this review, we highlight the emerging trends and future promises of immunotherapy for brain tumors and discuss PDT-OBBB as a new niche and an important informative platform for the development of innovative pharmacological strategies for the modulation of brain tumor immunity and the improvement of immunotherapy for GBM.

1. Photodynamic Therapy of Glioblastoma

Worldwide, brain tumors have been diagnosed in 300,000 people, among whom 250,000 patients have died [1,2]. In 2000, the 5- and 10-year survival rates for aggressive brain tumors were approximately 12% and 9%, respectively. Currently, this situation has improved, and the 5- and 10-year survival rates for malignant brain cancers are approximately 36% and 31%, respectively [3]. However, despite the improvements in therapy for brain tumors, the survival rate is still low compared to other types of cancer [1].
Glioblastoma (GBM) is the most common and aggressive forms of brain cancer [4,5]. GBM remains the one of deadliest of brain tumors. A guided surgical resection is the first step in treating patients with GBM [6,7]. However, even if GBM resection is successfully performed, 80% of GBMs recur within 2 cm of the original lesion [8,9,10]. There is no standard treatment for relapse. Surgery, radiation therapy, and chemotherapy or systemic therapy with bevacizumab are all options, depending on patient circumstances [11]. Typically, a patient who has surgery will receive chemotherapy, which is often combined with radiation [10]. However, despite all these treatments, the median survival time (MST) for most patients with GBM is less than one year, with a 5-year survival rate of less than 7% [12,13]. There are several challenges limiting the effective treatment of GBM [8]. First, patients can have an inoperable GBT due to tumor location, e.g., in the functional centers, which involve language, senses, vision and memory [14,15]. Second, the blood–brain barrier (BBB), due to its semipermeable properties, limits the delivery of a vast majority of cancer therapeutics, including monoclonal antibodies, antibody-drug conjugates, and hydrophilic molecules [16]. Third, GBM is characterized by heterogeneity at the cellular level, which induces different responses to anti-tumor pharmacological agents, leading to the failure of targeted therapies [17,18]. The challenge for GBM therapy is, therefore, to improve the control of tumor growth, especially for non-surgical patients.
Over the past 20 years, the innovative technology known as photodynamic therapy (PDT) has been developed for the treatment of GBM [19,20,21,22,23,24,25,26,27,28,29,30]. There are two modalities of PDT: intracavitary PDT and interstitial PDT (iPDT) (Figure 1). Intracavitary PDT is performed after the surgical resection of primary GBM with the use of an expandable balloon illumination device insufflated inside the surgical cavity [22,23]. The iPDT modality is a minimally invasive treatment for GBM without the large craniotomy that is used for non-surgical lesions or for therapy for recurrent GBM. iPDT is delivered with semiconductor lasers positioned inside the surgical cavity or directly in the tumor mass [20,24]. PDT combines a light source and nontoxic photosensitizing agents (photosensitizers, PSs). Systematically or topically administered PS is specifically accumulated in tumor tissues. Currently, the United States Food and Drug Administration (FDA) approved 5-aminolevulinic acid (5-ALA) for the fluorescence guided resection of GBMs and PDT of brain tumors [25]. 5-ALA exhibits high selectivity in terms of preferential accumulation in GBM tissues [26]. When the concentration of PS is sufficient, it is activated by exposure to light appropriated for its excitation (e.g., protoporphyrin IX (PpIX) induced by 5-ALA after its excitation with a 635 nm laser). The excited PS stimulates an energy transfer to endogenous oxygen (triplet state), leading to the generation of a singlet oxygen and resulting in tissue oxidation. In the singlet state, energy is converted to heat by internal conversion or emitted as fluorescence. In the triplet state, the energy generates the ROS-inducing damage within tumor cells by direct injury via necrosis and apoptosis or due to the occlusion of tumor vessels (thrombus formation) [19] (Figure 1). There are several clinical reports suggesting that PDT is a promising add-on therapy to the current standard of care for patients with GBM, especially in its recurrent and unresectable forms [19,20,21,22,23,24,25,26,27,28,29,30]. In the last 10 years, new PDT technologies have been developed based on the use of new-generation lasers capable of generating a singlet oxygen without PS [31,32,33]. PS-free-PDT may become a promising therapeutic approach in developing a breakthrough technology for the non-invasive treatment of GBM in children in whom PDT and radio- and chemotherapy are strongly limited, as well as in adult patients with allergic reactions to PSs. It should also be emphasized that children often show a superficial location of GBM in the brain [34,35], making the non-invasive treatment of pediatric tumors with PS-free-PDT possible.
The traditional explanation of the anti-cancer PDT effect is that the single-oxygen-induced damage of the endothelial cells results in tumor cell death and microvasculature collapse [19,20,21,27]. However, in recent studies, the new vascular effects of PDT associated with the opening of the BBB and the activation of lymphatic drainage of the brain tissues have been discovered [36,37,38,39,40,41,42,43,44]. In the next sections of this review, we discuss the mechanisms of PDT-OBBB and the PDT-OBBB-mediated modulation of brain tumor immunity (Figure 2).

2. Photodynamic Opening of the Blood–Brain Barrier

The BBB plays an important role in the regulation of CNS health and protects the CNS from toxins and pathogens [45]. Due to these properties, the BBB limits the delivery of a vast majority of anti-tumor therapeutics [15]. However, the question of whether the BBB actually prevents drug delivery into a GBT remains controversial [46]. It well known that the BBB is highly permeable in the mass of a GBM [47,48,49]. Nevertheless, the GBM cells spread via the blood vessels with an intact BBB [46,47]. Therefore, an effective GBM therapy depends on the development of new strategies to prevent the migration of the GBT cells via healthy cerebral vessels with an intact BBB [46,49,50,51].
In 2008, it was revealed that PDT, as an alternative method for GBM therapy, can also open the BBB [36,37,38,39,40,41,42,43,44]. Hirschberg was the first to publish the results of a study using inbred male Fischer 344 rats that showed that ALA-mediated PDT (400 µm bare flat-end quartz fiber (635 nm)) causes OBBB [36]. The BBB was opened 2 h after PDT treatment and completely restored 72 h after laser irradiation. No effect was observed on the BBB in the absence of 5-ALA. These findings clearly demonstrate PDT dose-related changes in rat brains. At the low fluence level of 9 J/cm2, 5-ALA-mediated PDT opened the BBB without any damages to brain tissues, with fast restoration of BBB permeability. At the high fluence level of 26 J/cm2, 5-ALA-mediated PDT caused both BBB disruption and damage to surrounding tissues (necrosis, edema, and hemorrhages), which were observed during the 2 weeks after 5-ALA-mediated PDT treatment.
In our experiments with 103 mongrel male mice, using different doses of laser radiation (635 nm, 10–40 J/cm2) and 5-ALA (20 and 80 mg/kg, i.v.), we revealed the optimal doses of PDT-OBBB (laser dose—15 J/cm2 and 5-ALA—20 mg/kg) for the fast recovery of the BBB after PDT treatment [42]. The higher doses of laser radiation or 5-ALA had no amplifying effects on BBB permeability, but they were associated with severe damage to the brain tissues. Zhang et al., using the method of skull optical clearing (USOCA [52]), showed non-invasive PDT-OBBB for an albumin complex of Evans Blue dye 68.5 kDa, TRITC dextran 70 kDa, and 100 nm GM1-liposomes in BALB/c mice [41]. Later, this group demonstrated that the PDT-OBBB-related levels of high weight molecules and solutes was more pronounced in 4-week-old mice than in 8-week-old BALB/c animals [40]. They also developed a technology for the quantitative and qualitative assessment of PDT-OBBB to Evans Blue based on spectral imaging through the optical clearing skull window [44].
Matsen et al. showed the efficiency of PDT-OBBB using macrophages loaded with gold-nanoparticles to improve the photothermal therapy (PTT) of GBM [39]. To study the efficiency of the PDT-OBBB, they used a photosensitizer, aluminium phthalocyanine disulfonate (AlPcS2a, 670 nm), along with a light dose of 2.5 J/cm2) and iron oxide nanoparticle (120–180 nm)-loaded exogenous macrophages as a contrast agents for the magnetic resonance imaging of the BBB’s permeability [37,38]. The accumulation of iron oxide-loaded macrophages was observed in both the MRI scans and histological sections of the non-tumor bearing rat brains following PDT-OBBB.
Trinidad et al., using a human FaDu cancer cell line (head and neck squamous cell carcinoma), demonstrated a significant synergy effect of combining PDT (AlPcS2a-PDT at a fluence level of 0.25 J/cm2) and PTT (14 W/cm2 irradiance) compared to applying each modality separately [53].
The mechanisms responsible for PDT-OBBB-induced changes remain poorly understood. In an experiment on mice, using ex vivo confocal microscopy, it was found that PDT-OBBB caused a temporal decrease in the intensity of the signals from the tight junctions (TJ), such as claudin-5 (CLDN-5) and VE-cadherin, as the main components of BBB integrity [41]. This could be explained by the temporal loss of TJ on the surface of the cerebral endothelium due to possible activation of the internalization of TJ [41]. Indeed, PDT-OBBB induced an elevation of the arrestin beta-1 (ARRB1) level in the brain tissues that is responsible for starting the internalization process [54]. It was shown in an in vitro model of the BBB that an increase in BBB permeability is associated with the ARRB2-mediated internalization of the VE-cadherin [55]. PDT has a direct effect on increasing the gaps of the TJs between the cerebral endothelial cells via changes of cytoskeleton and vascular tone loss due to microtubule depolarization [56,57]. One mechanism of PDT-OBBB may be an imbalance in the control of aquaporin water channels in the astrocytes [58,59,60,61]. This hypothesis is based on experimental results suggesting a relationship between PDT-OBBB and the development of vasogenic edema after the use of different photosensitizers [41,60,61]. Oxidative stress is another possible mechanism underlying PDT-OBBB, as evidenced by the increase in malondialdehyde levels in the mouse brains [41,62].
Thus, PDT-OBBB is an important and informative platform for better understanding the mechanisms underlying the cerebrovascular effects of fluorescence-guided resections of brain tumors and for improving the guidelines for the PDT treatment of GMB.

3. PDT-OBBB Modulation of Brain Tumor Immunity

Recently, it was discovered that GBM is characterized by the reduced outflow of cerebrospinal fluid and that the meningeal lymphatic vessels (MLVs) regulate brain tumor drainage and immunity [63,64,65,66,67]. This new knowledge about the functions of MLVs opens new strategies in the stimulation of efficient immune responses against gliomas [64,68]. Hu et al. showed that the dorsal MLVs underwent extensive remodeling in mice with intracranial glioma-grafted GL261 cells. They revealed changes in the gene that controlled MLV remodeling, fluid drainage, and inflammatory and immunological responses in mice with glioma [64]. Notably, the mice with glioma and an overexpression of vascular endothelial growth factor C (VEGF-C) displayed better responses to the anti-tumor therapies, including combinations of the anti- programmed death-1 protein (PD1) and cytotoxic T-lymphocyte-associated protein 4 (CTLA-4) blockade. However, the blockade of key lymphatic proteins abolished this effect [64]. Indeed, the mice with glioma that were treated with lymphatic protein blockers, such as chemokine (C-C motif) ligand 21 (CCL21) and C chemokine receptor type 7 (CCR7), did not demonstrate improvements after receiving this anti-tumor therapy. These pioneering data suggest that VEGF-C potentiates checkpoint therapy via the CCL21/CCR7 pathway. Song et al. discovered that VEGF receptor-3 (VEGFR3) enhanced immune surveillance from GBM and improved the effectiveness of anti-tumor therapies with checkpoints inhibitors [69]. Both Hu et al. [64] and Song et al. [69] documented an important role of the MLVs in the regulation of brain tumor immunity in mice. Thus, the brain local delivery of VEGF-C can improve the anti-tumor effects of immune checkpoint inhibitors and enhance the presence of tumor-infiltrating lymphocytes in the microenvironment of GBM [63,70,71,72].
OBBB is accompanied by the rapid activation of lymphatic drainage of the brain tissues and clearance of unnecessary molecules from the CNS [43,73,74,75,76]. Using two methods for OBBB—loud music or sound and PDT—we demonstrated that OBBB is associated with the activation of the lymphatic clearance of different tracers (Evans blue dye, dextran 70 kDa, beta-amyloid, and gold nanorods) that crossed OBBB (from the brain and its removal into the deep cervical lymph nodes (dcLNs)) [43,73,74,75,76]. The activation of the clearance of substances from the brain after OBBB can be explained by the hypothesis of Monro and Kellie [77]. According to their hypothesis, the cerebral spinal, extracellular, and intestinal fluids create a state of volume equilibrium. Therefore, any increase in the volume of one of these fluids should be compensated by a brain drainage, i.e., the removal of fluids from the brain. OBBB causes a fluid influx from the bloodstream into the perivascular spaces that, in the case of PDT-OBBB, can induce vasogenic edema [60,61]. We hypothesized that OBBB stimulates the drainage of the brain tissues in a way that maintains extracellular homeostasis.
The study of the photo-modulation of MLVs and the mechanisms underlying this phenomenon is still in its infancy. The changes in the MLVs associated with OBBB may be explained by the expression of a number of similar proteins in the endothelium of lymphatic and blood vessels. Indeed, the lymphatic vessels are composed of the TJ proteins typical for the blood endothelium, such as claudin-5, a family of ZO proteins, and endothelial selective adhesion molecule; VE-cadherin; junctional adhesion molecule (JAM); and PECAM-1/CD31 [78,79]. In various series of our experimental studies, we showed that PDT-OBBB caused temporal changes in TJs expression in both the blood and lymphatic endothelium [41,75,80].
The PDT-mediated activation of lymphatic functions can be also explained by a PTD-induced activation of endothelial nitric oxide (NO) synthase [81]. The NO causes the relaxation of the blood and lymphatic endothelium via the opening of calcium (Ca2+)-activated potassium channels that lead to the dilation of the blood and lymphatic vessels [82]. In series of experiments using the PS-free generation of a singlet oxygen by a 1267 nm laser, it was discovered an important role of NO in the photo-induced dilation of the MLVs led to an increase in the lymphatic evacuation of red blood cells (RBCs) from mouse brains with intraventricular hemorrhages [83]. The non-specific blockade of the NO synthases inhibited the photo-mediated relaxation of the MLVs and the evacuation of RBCs from the brain into the dcLNs [83]. We assumed that PDT’s effects on the MLVs facilitated the lymphatic drainage of the brain tissues and the removal of cells/macromolecules from the CNS to the dcLNs.
Another mechanism of the singlet-oxygen-mediated control of vascular tone is the ability of a singlet oxygen to regulate endothelial relaxation [84]. The singlet oxygen induces the oxidation of the amino acid tryptophan in mammalian tissues, which leads to the cellular production of metabolites, such as N-formylkynurenine, with the activation of the haem-containing enzyme indoleamine 2,3-dioxygenase 1. This enzyme is widely expressed in the blood and lymphatic endothelium and contributes to the relaxation of vascular tone [84,85,86]. It was recently discovered that endothelial indoleamine 2,3-dioxygenase 1 caused the dilation of blood vessels via the formation of a singlet oxygen [84]. These pioneering findings shed light on the roles of singlet oxygen in the regulation of vascular tone, opening new perspectives on the modulation of systemic inflammatory responses of vasculature.
PDT has several unique properties that induce immunogenic cell death (ICD) [87,88,89,90,91,92,93,94,95,96,97]. Among the different mechanisms of PDT-induced ICD is the damage-associated molecular patterns (DAMPs) released from tumor cells caused by PDT [98,99,100]. In turn, DAMPs mediate the formation of antigen-presenting cells (APCs). The PDT activation of APCs causes their migration and proliferation in local lymph nodes where the APCs then present tumor antigens to th CD8+ T cells [87]. The MLVs are pathways for APC traffic from the CNS into the dcLNs [63,69,70,71]. The activated CD8+ T cells induce the apoptosis of tumor cells, thereby providing long-term tumor control. Therefore, PDT-induced ICD has the potential to stimulate immune activation, contributing to long-term tumor control [87,88,89,90,91,92,93,94,95,96,97]. However, the efficiency of PDT-induced ICD is reduced in a hypoxic tumor microenvironment, which limits the antitumor effects during PDT-related immunostimulation [101]. There is an emerging trend of using PDT-induced ICD in combination with nanotechnology to overcome this limitation [88,89,93,94,97]. The growing body of evidence indicates that manganese dioxide and catalase can stimulate the production of O2 via the catalysis of hydrogen peroxidase [102,103]. Therefore, nanocarries loading with manganese dioxide, catalase, and other oxygen-boosted PDTs are considered to increase tumor ICD [88,94,102,103]. Furthermore, perfuorocarbons and hemoglobin are used for the direct delivery of O2 in tumor hypoxic microenvironments [94]. In addition, another limitation of PDT is the low level of ICD induced by PDT, which is not enough for effective antitumor effects [97]. To solve this problem, a combination of PDT-induced ICD with PTT and chemotherapy has been proposed to increase the effectiveness of tumor ICD [94]. Currently, the control of the escape of immune cells into the tumor microenvironment using inhibitors of immune checkpoints is considered as a most promising strategy in antitumor therapies [104,105,106,107,108,109]. The luck of signaling molecules in tumors that can activate the generation of T cells in the lymphoid organs is one reason that only few numbers of effector T cells can enrich the tumor region, which allows tumor cells to evade immune surveillance [104,105]. Moreover, checkpoints generally inhibit the activated T cells after antigen recognition, leading to a decrease in antitumor immunity [105,106,107,108]. Therefore, in the past decade, the use of immune checkpoint blockers, such as CTLA4, PD-1, programmed death-ligand 1, and indoleamine 2,3-dioxygenase, has been proposed for the improvement of antitumor therapies [105,106,107,108,109,110,111,112]. The combined antitumor therapy of PDT-induced ICD and inhibitors of immune checkpoints is expected to be a new and promising strategy in tumor immunotherapy, including GBM treatment [87,94].
PDT-treated GBM cells may also act as a therapeutic anti-tumor vaccine [87,113]. In earlier studies, Etminan et al. demonstrated that the ALA-PDT of GBM cells promoted the attraction of dendritic cells and stimulated their maturation [114]. Indeed, co-cultured ALA-PDT-treated GBM cells with DCs induced the expression of CD83, a marker for the maturation of DCs, as well as the expression of stimulatory factors, such as CD40, CD80, and CD86. Photofrine-PDT-treated C6 glioma cells co-cultured with dendritic cells caused more effective DC differentiation, with a high expression of CD80 and major histocompatibility complex II [115]. The combination of Hypericin-mediated PDT with DCs in a high-grade glioma mouse model demonstrated the significant suppression of the growth of glioma cells [116]. An in vivo brain tumor model using F98 glioma cells has shown a systemic immune response generated by disulfonated aluminum phtalocyanine-mediated PDT co-cultured with macrophages [113]. Taking into account the limitations of light penetration into the brain, the further detailed studies of the effectiveness of PDT-produced vaccines for GBM in humans appears to be the most attractive approach. A better understanding of the activation of the antitumor immunity induced by PDT-produced vaccines would allow researchers to define the optimal protocols of the stimulation of the immune system to recognize and prevent the recurrence of GBM after surgery. The combination of PDT-induced anti-tumor vaccines with check-point inhibitors is an exciting field to explore.

4. Limitations of PDT in Therapy of GBT

PDT is a treatment modality which consists of light, oxygen, and PSs, and all these components have limitations [117,118,119,120]. The penetration of light depends on the optical properties of the head and brain tissues and the laser wavelength used for the PDT (Figure 3). There exists a heterogeneity between and within a tissue, leading to light scattering and a significant loss in the light energy that can reach the brain tissues. The brain fluids absorb light at longer wavelengths, and endogenous dyes, such as hemoglobin and melanin, absorb light at shorter wavelengths, which changes the light penetration depth into the brain tissues. Therefore, the photo-therapeutic window is between 600 and 1300 nm [121]. The light within a range of wavelengths between 620 and 850 nm has the most penetrating capability to achieve the maximum skin permeability [121,122,123] (Table 1). It is believed that light above 850 nm cannot generate a sufficient energy transfer to its triplet state to produce a singlet oxygen. However, in recent studies, it has been discovered that a 1267 nm light can generate singlet oxygen directly in different tissues, including the brain [31,32,33,124,125,126,127]. This discovery can open new horizons for developing PSs-free photo-therapies for GBM for patients that cannot use PDT due to allergies, age, and other limitations.
The 1O2 oxygen is another fundamental element of PDT. Indeed, the therapeutic efficacy of PDT strongly depends on the 1O2 content in the GBM cells. On the other hand, the GBM tissues are deprived of 1O2 as a result of the rapid growth of GBM cells and insufficient vasculature [128]. Furthermore, photosensitization itself quickly depletes cellular 1O2 levels in tumor cells. Therefore, the light dose has to be carefully adjusted and the light should be introduced in pulses [129]. For this reason, PDT is considered a self-limiting modality which causes its own inhibition. In order to overcome this problem, oxygen-boosted PDT is considered to increase the efficacy of PDT for the treatment of GBM [88,94,102,103].
PS is a key component of PDT. The PS accumulated in tumor cells is activated by a light with specific intensity and wavelength. The activated PS performs a photodynamic response, which underlies the anti-tumor effects of PDT. 5-ALA is the most widely used agent for the fluorescence-guided surgery of GBM, with further PDT the edge of the tumor [130]. It is generally accepted that 5-ALA accumulates directly in the GBM cells [26,131,132]. However, as discussed above, PDT with 5-ALA affects also the normal cerebral vessels, inducing OBBB in the different regions of the brain, which may be one reason for the vasogenic edema observed after 5-ALA-mediated PDT [36,37,38,39,40,41]. This provides a basis for revisiting our knowledge of the vascular effects of 5-ALA-mediated PDT for the optimization of the photo-diagnosis of and therapies for GBM.
Table 1. Depth of the penetration of light into the head and brain tissues in humans.
Table 1. Depth of the penetration of light into the head and brain tissues in humans.
First Author, YearTissues, mm/Laser, nmPenetration Depth (mm) and Transmittance (%)
Wan et al., 1981 [133]Scalp + Skull, 9–13 mm
400 nm10−5–10−4%
546 nm2 × 10−4–8 × 10−4%
630 nm2 × 10−3–9 × 10−3%
664 nm2 × 10−2%
703 nm3–10−2%
856 nm2 × 10−2–8 × 10−2%
Lychagov et al., 2006 [134]Skull, 4–14 mm, 810 nm1–16%
Scalp and skull, 7–20 mm, 810 nm0.5–5%
Svaasand and Ellingsen, 1983 [135] ♂ Neonatal brain
♀ Adult brain
488 nm♂ 1.3 mm ♀ 0.4 mm
514 nm♂ 1.1 mm ♀ 0.4 mm
660 nm♂ 3.7 mm ♀ 1.2 mm
1060 nm♂ 7.1 mm ♀ 3.2 mm
Stolik et al., 2000 [136]Adult brain
632 nm0.92 mm
675 nm1.38 mm
780 nm2.17 mm
835 nm2.52 mm
Jagdeo et al., 2012 [137]Skull 5 mm
633 mmLeft parietal lobe 3%
Right parietal lobe 3.7%
Frontal lobe 1%
830 mmLeft parietal lobe 9%
Right parietal lobe 9.2%
Frontal lobe 5.9%
Yue et al., 2015 [138]Scalp, skull, and adult brain (total 60 mm)
850 nmParietal, occipital, temporal, and frontal lobes, 10−4–10−3%
Barett and Gonzalez-Lima, 2013 [139]Skull
1064 nmFrontal lobe 2%
On the whole, the choice of optimal combinations of PS dose, light sources, and treatment parameters is very important in order to achieve successful results in the PDT of GBM. The emerging evidence confirms that 5-ALA-mediated PDT is effective not only in the field of brain oncology, but also in many other areas of medicine where surgery is impossible [117]. It is expected that the coming years will be a time of observation of the development of new diagnostic and therapeutic strategies with the use of 5-ALA acid. In addition, PDT-OBBB as new method of brain drug delivery deserves greater capacity and investment in clinical trials, which would allow scientists to learn more about the true translational potential of PDT.

5. Conclusions

Recently, it has been discovered that PDT, as a promising method for the prevention of the post-operation recurrence of GBM, causes temporal OBBB. The PDT-OBBB sheds light on the innovative strategies for the modulation of brain tumor immunity (Figure 4). The new knowledge about PDT-OBBB associated with the activation of lymphatic drainage and clearance of the brain tissues opens a new niche in immunotherapy for brain tumors. PDT is used as a promising add-on therapy to the current standard of care for patients with GBM, especially in its recurrent and inoperable forms. PDT-OBBB activates the MLV functions, which are pathways of traffic for the APCs produced by GBM after PDT. Therefore, PDT-OBBB can potentially stimulate the lymphatic traffic of the APCs into the dcLNs where the APCs activate the tumor antigens to the CD4+/CD8+ T cells. The T-cells migrate from the dcLNs to the GBM and extravasate in the tumor tissues via OBBB. Thus, PDT-OBBB can facilitate an increase in the number of infiltrating lymphocytes and T-cells, which induces GBM cytotoxicity, releasing more GBM antigens and contributing to brain tumor immunity. The immunotherapy with VEGF-C can be significantly improved by PDT-OBBB via contributing to the delivery of VEGF-C into the GBM. Thus, PDT-OBBB may promote the effectiveness of immune checkpoint inhibitors which have been shown to enhance the number and function of tumor-infiltrating lymphocytes in GBM patients. Therefore, PDT-OBBB is a new and important informative platform for the development of innovative pharmacological strategies for the modulation of brain tumor immunity and for the improvement of immunotherapy in brain tumors.

Author Contributions

O.S.-G. and J.K. initiated and supervised this review article. O.S.-G. wrote the abstract, conclusion, and the “Photodynamic opening of the BBB” and “PDT-OBBB-modulation of brain tumor immunity” sections. A.T., V.T., A.K., I.B. and E.S. wrote the “Photodynamic therapy of GBM” section. E.S. prepared the figures. All authors have read and agreed to the published version of the manuscript.

Funding

O.S.-G., A.T., A.K., V.T., I.B., E.S. and J.K. were supported by RF Governmental Grant No. 075-15-2022-1094, grants from the RSF (nos. 20-15-00090 and 21-75-10088), a grant from RFBR (no. 20-015-00308-a), and the China-a 19-515-55016 grant.

Conflicts of Interest

The authors declare that they have no competing interest.

A List of Abbreviations

5-ALA5-Aminolevulinic acid
ARRB1Arrestin beta-1
CaCalcium
CNSCentral nervous system
CLDN-5Claudin-5
CCL-21Chemokine (C-C motif) ligand 21
CCR7C-such as C chemokine receptor type 7
CTLA-4Cytotoxic T lymphocyte associated protein 4
DAMPsDamage-associated molecular patterns
dcLNsDeep cervical lymph nodes
FDAFood and Drug Administration
FaDuCancer cell—hypopharyngeal carcinoma cell line
GM1Ganglioside (monosialotetrahexosylganglioside)
GBMGlioblastoma
GMPGuanosine monophosphate
HTPHeme-transport protein
ICDImmunogenic cell death
JJoule (unit of energy)
JAMJunctional adhesion molecule
LYVE-1Lymphatic vessel endothelial hyaluronan receptor 1
MaMacrophages
MLVsMeningeal lymphatic vessels
MSTMedian survival time
NONitric oxide
OBBBOpening of the blood–brain barrier
ROSReactive oxygen species
PECAM-1Platelet/endothelial cell adhesion molecule 1, also known as cluster of differentiation 31 (CD31)
PDTPhotodynamic therapy
iPDTInterstitial photodynamic therapy
PSsPhotosensitizers
PpIXProtoporphyrin IX
PD1programmed death-1 protein
PTTPhotothermal therapy
RNARibonucleic acid
TRITC-Dextran 70 kDaTetramethylrhodamine-isothiocyanate-dextran 70 kilodaltons
TJTight junctions
VEGFVascular endothelial growth factor
VEGF-CVascular endothelial growth factor C
VEGFR3Vascular endothelial growth factor receptor-3
VE-cadherinVascular endothelial cadherin
WWatt (unit of power)
ZO proteinsZonula occludens proteins

References

  1. Mitusova, K.; Peltek, O.O.; Karpov, T.E.; Muslimov, A.R.; Zyuzin, M.V.; Timin, A.S. Overcoming the blood–brain barrier for the therapy of malignant brain tumor: Current status and prospects of drug delivery approaches. J. Nanobiotechnol. 2022, 20, 412–463. [Google Scholar] [CrossRef] [PubMed]
  2. Brain, Central Nervous System. Available online: https://gco.iarc.fr/today (accessed on 11 April 2022).
  3. Ostrom, Q.T.; Cioffi, G.; Gittleman, H.; Patil, N.; Waite, K.; Kruchko, C.; Barnholtz-Sloan, J.S. CBTRUS statistical report: Primary brain and other central nervous system tumors diagnosed in the United States in 2012–2016. Neuro. Oncol. 2019, 21, v1–v100. [Google Scholar] [CrossRef]
  4. Ostrom, Q.T.; Cote, D.J.; Ascha, M.; Kruchko, C.; Barnholtz-Sloan, J.S. Adult glioma incidence and survival by race or ethnicity in the United States from 2000 to 2014. JAMA Oncol. 2018, 4, 1254–1262. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  5. Minniti, G.; Lombardi, G.; Paolini, S. Glioblastoma in elderly patients: Current management and future perspectives. Cancers 2019, 11, 336. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  6. Sun, R.; Cuthbert, H.; Watts, C. Fluorescence-guided surgery in the surgical treatment of gliomas: Past, present and future. Cancers 2021, 13, 3508. [Google Scholar] [CrossRef] [PubMed]
  7. Wang, L.M.; Banu, M.A.; Canoll, P.; Bruce, J.N. Rationale and clinical implications of fluorescein-guided supramarginal resection in newly diagnosed high-grade glioma. Front. Oncol. 2021, 11, 666734. [Google Scholar] [CrossRef]
  8. Davis, M.E. Glioblastoma: Overview of disease and treatment. Clin. J. Oncol. Nurs. 2016, 20, S2–S8. [Google Scholar] [CrossRef] [Green Version]
  9. Neira, J.A.; Ung, T.H.; Sims, J.S.; Malone, H.R.; Chow, D.S.; Samanamud, J.L.; Zanazzi, G.J.; Guo, X.; Bowden, S.G.; Zhao, B.; et al. Aggressive resection at the infiltrative margins of glioblastoma facilitated by intraoperative fuorescein guidance. J. Neurosurg. 2017, 127, 111–122. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  10. Thon, N.; Tonn, J.C.; Kreth, F.W. The surgical perspective in precision treatment of diffuse gliomas. Onco. Targets Ther. 2019, 12, 1497–1508. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  11. Tan, A.C.; Ashley, D.M.; Lopez, G.Y.; Malinzak, M.; Friedman, H.S.; Khasraw, M. Management of glioblastoma: State of the art and future directions. CA Cancer J. Clin. 2020, 70, 299–312. [Google Scholar] [CrossRef] [PubMed]
  12. Ostrom, Q.T.; Cioffi, G.; Waite, K.; Kruchko, C.; Barnholtz-Sloan, J.S. CBTRUS statistical report: Primary brain and other central nervous system tumors diagnosed in the United States in 2014–2018. Neuro. Oncol. 2021, 23, iii1–iii105. [Google Scholar] [CrossRef] [PubMed]
  13. Low, J.T.; Ostrom, Q.T.; Cioffi, G.; Neff, C.; Waite, K.A.; Kruchko, C.; Barnholtz-Sloan, J.S. 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, 9, 165–182. [Google Scholar] [CrossRef]
  14. Allemani, C.; Matsuda, T.; Di Carlo, V.; Harewood, R.; Matz, M.; Nikšić, M.; Bonaventure, A.; Valkov, M.; Johnson, C.J.; Estève, J.; et al. Global surveillance of trends in cancer survival 2000–14 (CONCORD-3): Analysis of individual records for 37,513,025 patients diagnosed with one of 18 cancers from 322 population-based registries in 71 countries. Lancet 2018, 391, 1023–1075. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  15. Löber-Handwerker, R.; Döring, K.; Bock, C.; Rohde, V.; Malinova, V. Defining the impact of adjuvant treatment on the prognosis of patients with inoperable glioblastoma undergoing biopsy only: Does the survival benefit outweigh the treatment effort? Neurosurg. Rev. 2022, 45, 2339–2347. [Google Scholar] [CrossRef] [PubMed]
  16. Omidi, Y.; Kianinejad, N.; Kwon, Y.; Omidian, H. Drug delivery and targeting to brain tumors: Considerations for crossing the blood-brain barrier. Expert Rev. Clin. Pharmacol. 2021, 14, 357–381. [Google Scholar] [CrossRef] [PubMed]
  17. Yesudhas, D.; Dharshini, S.A.P.; Taguchi, Y.-H.; Gromiha, M.M. Tumor heterogeneity and molecular characteristics of glioblastoma revealed by single-cell RNA-Seq data analysis. Genes 2022, 13, 428. [Google Scholar] [CrossRef] [PubMed]
  18. Couturier, C.P.; Ayyadhury, S.; Le, P.U.; Nadaf, J.; Monlong, J.; Riva, G.; Allache, R.; Baig, S.; Yan, X.; Bourgey, M.; et al. Single-cell RNA-seq reveals that glioblastoma recapitulates a normal neurodevelopmental hierarchy. Nat. Commun. 2020, 11, 3406. [Google Scholar] [CrossRef] [PubMed]
  19. Henderson, B.W.; Dougherty, T.J. How does photodynamic therapy works? Photochem. Photobiol. 1992, 55, 145–157. [Google Scholar] [CrossRef] [PubMed]
  20. Leroy, H.-A.; Baert, G.; Guerin, L.; Delhem, N.; Mordon, S.; Reyns, N.; Vignion-Dewalle, A.-S. Interstitial photodynamic therapy for glioblastomas: A standardized procedure for clinical use. Cancers 2021, 13, 5754. [Google Scholar] [CrossRef] [PubMed]
  21. Cramer, S.W.; Chen, C.C. Photodynamic therapy for the treatment of glioblastoma. Front. Surg. 2020, 6, 81. [Google Scholar] [CrossRef]
  22. Dupont, C.; Vermandel, M.; Leroy, H.; Quidet, M.; Lecomte, F.; Delhem, N.; Mordon, S.; Reyns, N. Intraoperative Photodynamic Therapy for Glioblastomas (INDYGO): Study protocol for a phase I clinical trial. Neurosurgery 2018, 84, E414–E419. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  23. Vermandel, M.; Dupont, C.; Lecomte, F.; Leroy, H.-A.; Tuleasca, C.; Mordon, S.; Hadjipanayis, C.G.; Reyns, N. Standardized intraoperative 5-ALA photodynamic therapy for newly diagnosed glioblastoma patients: A preliminary analysis of the INDYGO clinical trial. J. Neuro-Oncol. 2021, 152, 501–514. [Google Scholar] [CrossRef] [PubMed]
  24. Muragaki, Y.; Akimoto, J.; Maruyama, T.; Iseki, H.; Ikuta, S.; Nitta, M.; Maebayashi, K.; Saito, T.; Okada, Y.; Kaneko, S.; et al. Phase II clinical study on intraoperative photodynamic therapy with Talaporfin sodium and semiconductor laser in patients with malignant brain tumors: Clinical article. J. Neurosurg. 2013, 119, 845–852. [Google Scholar] [CrossRef] [PubMed]
  25. Hadjipanayis, C.G.; Stummer, W. 5-ALA and FDA approval for glioma surgery. J. Neurooncol. 2019, 141, 479–486. [Google Scholar] [CrossRef] [PubMed]
  26. Mahmoudi, K.; Garvey, K.L.; Bouras, A.; Cramer, G.; Stepp, H.; Jesu Raj, J.G.; Bozec, D.; Busch, T.M.; Hadjipanayis, C.G. 5-Aminolevulinic acid photodynamic therapy for the treatment of high-grade gliomas. J. Neurooncol. 2019, 141, 595–607. [Google Scholar] [CrossRef]
  27. Kaneko, S.; Fujimoto, S.; Yamaguchi, H.; Yamauchi, T.; Yoshimoto, T.; Tokuda, K. Photodynamic therapy of malignant gliomas. Prog. Neurol. Surg. 2018, 32, 1–13. [Google Scholar]
  28. Stepp, H.; Beck, T.; Pongraz, T.; Meinel, T.; Kreth, F.W.; Tonn, J.C.; Stummer, W. ALA and malignant glioma: Fluorescence-guided resection and photodynamic treatment. J. Environ. Pathol. Toxicol. Oncol. 2007, 21, 157–164. [Google Scholar] [CrossRef] [PubMed]
  29. Beck, T.J.; Kreth, F.W.; Beyer, W.; Mehrkens, J.H.; Obermeier, A.; Stepp, H.; Stummer, W.; Baumgartner, R. Interstitial photodynamic therapy of nonresectable malignant glioma recurrences using 5-aminolevulinic acid induced protoporphyrin IX. Lasers Surg. Med. 2007, 39, 386–393. [Google Scholar] [CrossRef]
  30. Schwartz, C.; Ruhm, A.; Tonn, J.-C.; Kreth, S.; Kreth, F.-W. Interstital photodynamic therapy of de-novo glioblastoma multiformeWHO IV. Neurooncology 2015, 17, 214–220. [Google Scholar]
  31. Sokolovski, S.G.; Zolotovskaya, S.A.; Goltsov, A.; Pourreyron, C.; South, A.P.; Rafailov, E.U. Infrared laser pulse triggers increased singlet oxygen production in tumour cells. Sci. Rep. 2013, 3, 3484. [Google Scholar] [CrossRef]
  32. Khokhlova, A.; Zolotovskii, I.; Sokolovski, S.; Saenko, Y.; Rafailov, E.; Stoliarov, D.; Pogodina, E.; Svetukhin, V.; Sibirny, V.; Fotiadi, A. The light-oxygen effect in biological cells enhanced by highly localized surface plasmon-polaritons. Sci. Rep. 2019, 9, 18435. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  33. Khokhlova, A.; Zolotovskii, I.; Stoliarov, D.; Vorsina, S.; Liamina, D.; Pogodina, E.; Fotiadi, A.A.; Sokolovski, S.G.; Saenko, Y.; Rafailov, E.U. The photobiomodulation of vital parameters of the cancer cell culture by low dose of near-IR laser Iirradiation. IEEE J. Sel. Top. Quantum Electron. 2019, 25, 1–10. [Google Scholar] [CrossRef]
  34. Koeller, K.K.; Henry, J.M. From the archives of the AFIP: Superficial gliomas: Radiologic-pathologic correlation. Armed Forces Institute of Pathology. Radiographics 2001, 21, 1533–1556. [Google Scholar] [CrossRef] [PubMed]
  35. Blionas, A.; Giakoumettis, D.; Klonou, A.; Neromyliotis, E.; Karydakis, P.; Themistocleous, M.S. Paediatric gliomas: Diagnosis, molecular biology and management. Ann. Transl. Med. 2018, 6, 251. [Google Scholar] [CrossRef]
  36. Hirschberg, H. Disruption of the blood–brain barrier following ALA-mediated photodynamic therapy. Lasers Surg. Med. 2008, 40, 535–542. [Google Scholar] [CrossRef] [Green Version]
  37. Madsen, S.J. Site-specific opening of the blood-brain barrier. J. Biophoton. 2010, 3, 356–367. [Google Scholar] [CrossRef] [Green Version]
  38. Madsen, S.J. Increased nanoparticle-loaded exogenous macrophage migration into the brain following PDT-induced blood-brain barrier disruption. Lasers Surg. Med. 2013, 45, 524–532. [Google Scholar] [CrossRef] [Green Version]
  39. Madsen, S.J. Nanoparticle-loaded macrophage mediated photothermal therapy: Potential for glioma treatment. Lasers Med. Sci. 2015, 4, 1357–1365. [Google Scholar] [CrossRef] [Green Version]
  40. Zhang, C.; Feng, W.; Li, Y.; Kürths, J.; Yu, T.; Semyachkina-Glushkovskaya, O.; Zhu, D. Age differences in photodynamic opening of blood-brain barrier through optical clearing skull window in mice. Lasers Surg. Med. 2019, 51, 625–633. [Google Scholar] [CrossRef]
  41. Zhang, C.; Feng, W.; Vodovosova, E.; Tretiakova, D.; Boldyrev, I.; Li, Y.; Kurths, J.; Yu, T.; Semyachkina-Glushkovskaya, O.; Zhu, D. Photodynamic opening of the blood-brain barrier to high weight molecules and liposomes through an optical clearing skull window. Biomed. Opt. Express 2018, 9, 4850–4862. [Google Scholar] [CrossRef]
  42. Semyachkina-Glushkovskaya, O.; Kurths, J.; Borisova, E.; Sokolovsky, S.; Mantareva, N.; Angelov, I.; Shirokov, A.; Navolokin, N.; Shushunova, N.; Khorovodov, A.; et al. Photodynamic opening of blood-brain barrier. Biomed. Opt. Express 2017, 8, 5040–5048. [Google Scholar] [CrossRef] [PubMed]
  43. Semyachkina-Glushkovskaya, O.; Chehonin, V.; Borisova, E.; Fedosov, I.; Namykin, A.; Abdurashitov, A.; Shirokov, A.; Khlebtsov, B.; Lyubun, Y.; Navolokin, N.; et al. Photodynamic opening of the blood-brain barrier and pathways of brain clearing pathways. J. Biophotonics 2018, 11, e201700287. [Google Scholar] [CrossRef] [PubMed]
  44. Feng, W.; Zhang, C.; Yu, T.; Semyachkina-Glushkovskaya, O.; Zhu, D. In vivo monitoring blood-brain barrier permeability using spectral imaging through optical clearing skull window. J. Biophotonics 2019, 12, e201800330. [Google Scholar] [CrossRef] [PubMed]
  45. Abbott, N.J.; Patabendige, A.A.K.; Dolman, D.E.M.; Yusof, S.R.; Begley, D.J. Structure and function of the blood–brain barrier. Neurobiol. Dis. 2010, 37, 13–25. [Google Scholar] [CrossRef] [PubMed]
  46. Sarkaria, J.N.; Hu, L.S.; Parney, I.F.; Pafundi, D.H.; Brinkmann, D.H.; Laack, N.N.; Giannini, C.; Burns, T.C.; Kizilbash, S.H.; Laramy, J.K.; et al. Is the blood-brain barrier really disrupted in all glioblastomas? A critical assessment of existing clinical data. Neuro. Oncol. 2018, 20, 184–191. [Google Scholar] [CrossRef]
  47. Arvanitis, C.D.; Ferraro, G.B.; Jain, R.K. The blood-brain barrier and blood-tumour barrier in brain tumours and metastases. Nat. Rev. Cancer 2020, 20, 26–41. [Google Scholar] [CrossRef]
  48. Dubois, L.G.; Campanati, L.; Righy, C.; D’Andrea-Meira, I.; Leite de Sampaio e Spohr, T.C.; Porto-Carreiro, I.; Pereira, C.M.; Balça-Silva, J.; Assad Kahn, S.; DosSantos, M.F.; et al. Gliomas and the vascular fragility of the blood brain barrier. Front. Cell. Neurosci. 2014, 8, 418. [Google Scholar] [CrossRef] [Green Version]
  49. Noell, S.; Ritz, R.; Wolburg-Buchholz, K.; Wolburg, H.; Fallier-Becker, P. An allograft glioma model reveals the dependence of aquaporin-4 expression on the brain microenvironment. PLoS ONE 2012, 7, e36555. [Google Scholar] [CrossRef] [Green Version]
  50. Wang, D.; Wang, C.; Wang, L.; Chen, Y. Comprehensive review in improving delivery of small-molecule chemotherapeutic agents overcoming the blood–brain/brain tumor barriers for glioblastoma treatment. Drug Deliv. 2019, 26, 551–565. [Google Scholar] [CrossRef]
  51. Agarwal, S.; Sane, R.; Oberoi, R.; Ohlfest, J.R.; Elmquist, W.F. 2011 Delivery of molecularly targeted therapy to malignant glioma, a disease of the whole brain. Expert Rev. Mol. Med. 2011, 13, 17. [Google Scholar] [CrossRef] [Green Version]
  52. Zhang, C.; Feng, W.; Zhao, Y.; Yu, T.; Li, P.; Xu, T.; Luo, Q.; Zhu, D. A large, switchable optical clearing skull window for cerebrovascular imaging. Theranostics 2018, 8, 2696–2708. [Google Scholar] [CrossRef] [PubMed]
  53. Trinidad, J.; Hong, S.J.; Peng, Q.; Madsen, S.J.; Hirschberg, H. Combined concurrent photodynamic and gold nanoshell loaded macrophagemediated photothermal therapies: An in vitro study on squamous cell head and neck carcinoma. Lasers Surg. Med. 2014, 4, 310–318. [Google Scholar] [CrossRef] [PubMed]
  54. Hara, M.R.; Kovacs, J.J.; Whalen, E.J.; Rajagopal, S.; Strachan, R.T.; Grant, W.; Towers, A.J.; Williams, B.; Lam, C.M.; Xiao, K.; et al. A stress response pathway regulates DNA damage through β2-adrenoreceptors and β-arrestin-1. Nature 2011, 477, 349–353. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  55. Hebda, J.K.; Leclair, H.M.; Azzi, S.; Roussel, C.; Scott, M.G.; Bidère, N.; Gavard, J. The C-terminus region of β-arrestin1 modulates VE-cadherin expression and endothelial cell permeability. Cell Commun. Signal. 2013, 11, 37. [Google Scholar] [CrossRef] [Green Version]
  56. Hu, S.S.; Cheng, H.B.; Zheng, Y.R.; Zhang, R.Y.; Yue, W.; Zhang, H. Effects of photodynamic therapy on the ultrastructure of glioma cells. Biomed. Environ. Sci. 2007, 20, 269–273. [Google Scholar]
  57. Buzza, H.; de Fraitas, L.C.F.; Moriayama, L.T.; Rosa, R.; Bagnato, F.; Kurachi, C. Vascular effects of photodynamic therapy with circumin in a chlorioallantoic membrane model. Int. J. Mol. Sci. 2019, 20, 1084. [Google Scholar] [CrossRef] [Green Version]
  58. Filippidis, S.; Carozza, R.B.; Rekate, H.L. Aquaporins in brain edema and neuropathological conditions. Int. J. Mol. Sci. 2017, 18, 55. [Google Scholar] [CrossRef] [Green Version]
  59. Clément, T.; Rodriguez-Grandel, B.; Badaut, J. Aquaporins in brain edema. J. Neurosci. Res. 2020, 98, 9–18. [Google Scholar] [CrossRef] [Green Version]
  60. Ito, S.; Rachinger, W.; Stepp, H.; Reulen, H.J.; Stummer, W. Oedema formation in experimental photoirradiation therapy of brain tumours using 5-ALA. Acta Neurochir. 2005, 147, 57–65. [Google Scholar] [CrossRef]
  61. Mathews, M.S.; Chighvinadze, D.; Gach, H.M.; Uzal, F.A.; Madsen, S.J.; Hirschberg, H. Cerebral edema following photodynamic therapy using endogenous and exogenous photosensitizers in normal brain. Lasers Surg. Med. 2011, 43, 892–900. [Google Scholar] [CrossRef] [Green Version]
  62. Dharmajaya, R.; Sari, D.K. Malondialdehyde value as radical oxidative marker and endogenous antioxidant value analysis in brain tumor. Ann. Med. Surg. 2022, 77, 103231. [Google Scholar] [CrossRef] [PubMed]
  63. Kanamori, M.; Kipnis, J. Meningeal lymphatics “drain” brain tumors. Cell Res. 2020, 30, 191–192. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  64. Hu, X.; Deng, Q.; Ma, L.; Li, Q.; Chen, Y.; Liao, Y.; Zhou, F.; Zhang, C.; Shao, L.; Feng, J.; et al. Meningeal lymphatic vessels regulate brain tumor drainage and immunity. Cell Res. 2020, 30, 229–243. [Google Scholar] [CrossRef] [PubMed]
  65. Ma, Q.; Schlegel, F.; Bachmann, S.B.; Hannah Schneider, H.; Decker, Y.; Rudin, M.; Weller, M.; Proulx, S.T.; Detmar, M. Lymphatic outflow of cerebrospinal fluid is reduced in glioma. Sci. Rep. 2019, 9, 14815. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  66. Norton, E.S.; Whaley, L.A.; Ulloa-Navas, M.J.; Patricia García-Tárraga, P.; Meneses, K.M.; Lara-Velazquez, M.; Zarco, N.; Carrano, A.; Quiñones-Hinojosa, A.; García-Verdugo, J.M.; et al. Glioblastoma disrupts the ependymal wall and extracellular matrix structures of the subventricular zone. Fluids Barriers CNS 2022, 19, 58. [Google Scholar] [CrossRef]
  67. Xu, D.; Zhou, J.; Mei, H.; Li, H.; Sun, W.; Xu, H. Impediment of cerebrospinal fluid drainage through glymphatic system in glioma. Front. Oncol. 2022, 11, 790821. [Google Scholar] [CrossRef]
  68. Lan, Y.L.; Wang, H.; Chen, A.; Zhang, J. Update on the current knowledge of lymphatic drainage system and its emerging roles in glioma management. Immunology 2022, 1–15. [Google Scholar] [CrossRef]
  69. Song, E.; Mao, T.; Dong, H.; Boisserand, L.S.B.; Antila, S.; Bosenberg, M.; Alitalo, K.; Thomas, J.L.; Iwasaki, A. VEGF-C-driven lymphatic drainage enables immunosurveillance of brain tumours. Nature 2020, 577, 689–694. [Google Scholar] [CrossRef]
  70. Graham, M.S.; Mellinghoff, I.K. Meningeal lymphatics prime tumor immunity in glioblastoma. Cancer Cell 2021, 39, 304–306. [Google Scholar] [CrossRef]
  71. Bordon, Y. VEGF-C shines a light on brain tumours. Nat. Rev. Immunol. 2020, 20, 140–141. [Google Scholar] [CrossRef]
  72. Cloughesy, T.F.; Mochizuki, A.Y.; Orpilla, J.R.; Hugo, W.; Lee, A.H.; Davidson, T.B.; Wang, A.C.; Ellingson, B.M.; Rytlewski, J.A.; Sanders, C.M. Neoadjuvant anti-PD-1 immunotherapy promotes a survival benefit with intratumoral and systemic immune responses in recurrent glioblastoma. Nat. Med. 2019, 25, 477–486. [Google Scholar] [CrossRef] [PubMed]
  73. Semyachkina-Glushkovskaya, O.; Bragin, D.; Bragina, O.; Yang, Y.; Abdurashitov, A.; Esmat, A.; Khorovodov, A.; Terskov, A.; Klimova, M.; Agranovich, I.; et al. Mechanisms of sound-induced opening of the blood-brain barrier. Adv. Exp. Med. Biol. 2021, 1269, 197–202. [Google Scholar] [PubMed]
  74. Semyachkina-Glushkovskaya, O.; Esmat, A.; Bragin, D.; Bragina, O.; Shirokov, A.A.; Navolokin, N.; Yang, Y.; Abdurashitov, A.; Khorovodov, A.; Terskov, A.; et al. Phenomenon of music-induced opening of the blood-brain barrier in healthy mice. Proc. R. Soc. B 2020, 287, 20202337. [Google Scholar] [CrossRef] [PubMed]
  75. Semyachkina-Glushkovskaya, O.; Abdurashitov, A.; Klimova, M.; Dubrovsky, A.; Shirokov, A.; Fomin, A.; Terskov, A.; Agranovich, I.; Mamedova, M.; Khorovodov, A.; et al. Photostimulation of cerebral and peripheral lymphatic functions. Translat. Biophot. 2020, 2, e201900036. [Google Scholar] [CrossRef]
  76. Semyachkina-Glushkovskaya, O.; Khorovodov, A.; Fedosov, I.; Pavlov, A.; Shirokov, A.; Sharif, A.E.; Dubrovsky, A.; Blokhina, I.; Terskov, A.; Navolokin, N.; et al. A novel method to stimulate lymphatic clearance of beta-amyloid from mouse brain using noninvasive music-induced opening of the blood–brain barrier with EEG markers. Appl. Sci. 2021, 11, 10287. [Google Scholar] [CrossRef]
  77. Mokri, B. The Monro-Kellie hypothesis: Applications in CSF volume depletion. Neurology 2001, 56, 1746–1748. [Google Scholar] [CrossRef] [PubMed]
  78. Batuk, P.; Fuxe, J.; Hashizume, H.; Romano, T.; Lashnits, E.; Butz, S.; Vestweber, D.; Corada, M.; Molendinin, C.; Dejana, E. Functionally specialized junctions between endothelial cells of lymphatic vessels. J. Exp. Med. 2004, 204, 2349–2362. [Google Scholar]
  79. Kesler, C.; Kiao, S.; Munn, L.; Padera, T. Lymphatic vessels in health and diseases. Wiley Interdiscip. Rev. Syst. Biol. Med. 2013, 5, 111–124. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  80. Semyachkina-Glushkovskaya, O.; Borisova, E.; Mantareva, V.; Angelov, I.; Eneva, I.; Terskov, A.; Mamedova, A.; Shirokov, A.; Khorovodov, A.; Klimova, M.; et al. Photodynamic opening of the blood–brain barrier using different photosensitizers in mice. Appl. Sci. 2020, 10, 33. [Google Scholar] [CrossRef] [Green Version]
  81. Karu, T.I.; Pyatibrat, L.V.; Afanasyeva, N.I. Cellular effects of low power laser therapy can be mediated by nitric oxide. Lasers Surg. Med. 2005, 36, 307–314. [Google Scholar] [CrossRef] [PubMed]
  82. Murad, F. Discovery of some of the biological effects of nitric oxide and its role in cell signaling. Biosci. Rep. 2004, 24, 452–474. [Google Scholar] [CrossRef] [PubMed]
  83. Li, G.Y.; Liu, S.J.; Yu, T.T.; Liu, Z.; Sun, S.L.; Bragin, D.; Navolokin, N.; Kurths, J.; Glushkovskaya-Semyachkina, O.; Zhu, D. Photostimulation of lymphatic clearance of red blood cells from the mouse brain after intraventricular hemorrhage. bioRxiv 2020. [Google Scholar] [CrossRef]
  84. Stanley, C.P.; Maghzal, G.J.; Ayer, A.; Talib, J.; Giltrap, A.M.; Shengule, S.; Wolhuter, K.; Wang, Y.; Chadha, P.; Suarna, C.; et al. Singlet molecular oxygen regulates vascular tone and blood pressure in inflammation. Nature 2019, 566, 548–552. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  85. Ronsein, G.E.; Oliveira, M.C.; Miyamoto, S.; Medeiros, M.H.G.; Di Mascio, P. Tryptophan oxidation by singlet molecular oxygen [O2(1∆g)]: Mechanistic studies using 18O-labeled hydroperoxides, mass spectrometry, and light emission measurements. Chem. Res. Toxicol. 2008, 21, 1271–1283. [Google Scholar] [CrossRef] [PubMed]
  86. Nakagawa, M.; Kato, S.; Nakano, K.; Hino, T. Dye-sensitized photooxygenation of tryptophan: 3a-hydroperoxypyrroloindole as a labile precursor of formylkynurenine. Chem. Pharm. Bull. 1981, 29, 1013–1026. [Google Scholar] [CrossRef] [Green Version]
  87. Hirschberg, H.; Berg, K.; Peng, Q. Photodynamic therapy mediated immune therapy of brain tumors. Neuroimmunol. Neuroinflamm. 2018, 5, 27. [Google Scholar] [CrossRef] [PubMed]
  88. Alzeibak, R.; Mishchenko, T.A.; Shilyagina, N.Y. Targeting immunogenic cancer cell death by photodynamic therapy: Past, present and future. J. Immun. Ther. Cancer 2021, 9, e001926. [Google Scholar] [CrossRef] [PubMed]
  89. Rodrigues, M.C.; de Sousa Júnior, W.T.; Mundim, T.; Vale, C.L.C.; de Oliveira, J.V.; Ganassin, R.; Pacheco, T.J.A.; Vasconcelos Morais, J.A.; Longo, J.P.F.; Azevedo, R.B. Induction of Immunogenic Cell Death by Photodynamic Therapy Mediated by Aluminum-Phthalocyanine in Nanoemulsion. Pharmaceutics 2022, 14, 196. [Google Scholar] [CrossRef]
  90. Turubanova, V.D.; Mishchenko, T.A.; Balalaeva, I.V.; Efimova, I.; Peskova, N.N.; Klapshina, L.G.; Lermontova, S.A.; Bachert, C.; Krysko, O.; Vedunova, M.V.; et al. Novel porphyrazine-based photodynamic anti-cancer therapy induces immunogenic cell death. Sci. Rep. 2021, 11, 7205. [Google Scholar] [CrossRef] [PubMed]
  91. Morais, J.A.V.; Almeida, L.R.; Rodrigues, M.C.; Azevedo, R.B.; Muehlmann, L.A. The induction of immunogenic cell death by photodynamic therapy in B16F10 cells in vitro is effected by the concentration of the photosensitizer. Photodiagnosis Photodyn Ther. 2021, 35, 102392. [Google Scholar] [CrossRef] [PubMed]
  92. Mishchenko, T.; Balalaeva, I.; Gorokhova, A.; Vedunova, M.; Krysko, D.V. Which cell death modality wins the contest for photodynamic therapy of cancer? Cell Death Dis. 2022, 13, 455. [Google Scholar] [CrossRef] [PubMed]
  93. Zhang, S.; Wang, J.; Kong, Z.; Sun, X.; He, Z.; Sun, B.; Luo, C.; Sun, J. Emerging photodynamic nanotherapeutics for inducing immunogenic cell death and potentiating cancer immunotherapy. Biomaterials 2022, 282, 121433. [Google Scholar] [CrossRef] [PubMed]
  94. Wei, X.; Song, M.; Jiang, G.; Liang, M.; Chen, C.; Yang, Z.; Zou, L. Progress in advanced nanotherapeutics for enhanced photodynamic immunotherapy of tumor. Theranostics 2022, 12, 5272–5298. [Google Scholar] [CrossRef] [PubMed]
  95. Jin, F.; Liu, D.; Xu, X.; Ji, J.; Du, Y. Nanomaterials-Based Photodynamic Therapy with Combined Treatment Improves Antitumor Efficacy Through Boosting Immunogenic Cell Death. Int. J. Nanomed. 2021, 16, 4693–4712. [Google Scholar] [CrossRef] [PubMed]
  96. Song, H.; Cai, Z.; Li, J.; Xiao, H.; Qi, R.; Zheng, M. Light triggered release of a triple action porphyrin-cisplatin conjugate evokes stronger immunogenic cell death for chemotherapy, photodynamic therapy and cancer immunotherapy. J. Nanobiotech. 2022, 20, 329. [Google Scholar] [CrossRef] [PubMed]
  97. Zhang, G.; Wang, N.; Sun, H.; Fu, X.; Zhai, S.; Cui, J. Self-adjuvanting photosensitizer nanoparticles for combination photodynamic immunotherapy. Biomater. Sci. 2021, 9, 6940–6949. [Google Scholar] [CrossRef] [PubMed]
  98. Krysko, D.V.; Garg, A.D.; Kaczmarek, A.; Krysko, O.; Agostinis, P.; Vandenabeele, P. Immunogenic cell death and DAMPs in cancer therapy. Nat. Rev. Cancer 2012, 12, 860–875. [Google Scholar] [CrossRef]
  99. Kroemer, G.; Galluzzi, L.; Kepp, O.; Zitvogel, L. Immunogenic cell death in cancer therapy. Annu. Rev. Immunol. 2013, 31, 51–72. [Google Scholar] [CrossRef] [PubMed]
  100. Castano, A.P.; Mroz, P.; Hamblin, M.R. Photodynamic therapy and anti-tumour immunity. Nat. Rev. Cancer 2006, 6, 535–545. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  101. Li, X.; Kwon, N.; Gu, T.; Liu, Z.; Yoon, J. Innovative strategies for hypoxic-tumor photodynamic therapy. Angew. Chem. Int. Ed. 2018, 57, 11522–11531. [Google Scholar] [CrossRef] [PubMed]
  102. Li, Q.; Ren, J.; Chen, Q.; Liu, W.; Xu, Z.; Cao, Y. A HMCuS@MnO2 nanocomplex responsive to multiple tumor environmental clues for photoacoustic/fluorescence/magnetic resonance trimodal imaging-guided and enhanced photothermal/photodynamic therapy. Nanoscale 2020, 12, 12508–12521. [Google Scholar] [CrossRef] [PubMed]
  103. Cheng, H.; Zhu, J.-Y.; Li, S.-Y.; Zeng, J.-Y.; Lei, Q.; Chen, K.-W. An O2 self-sufficient biomimetic nanoplatform for highly specific and efficient photodynamic therapy. Adv. Funct. Mater. 2016, 26, 7847–7860. [Google Scholar] [CrossRef]
  104. Waldman, A.D.; Fritz, J.M.; Lenardo, M.J. A guide to cancer immunotherapy: From T cell basic science to clinical practice. Nat. Rev. Immunol. 2020, 20, 651–668. [Google Scholar] [CrossRef] [PubMed]
  105. Xia, A.; Zhang, Y.; Xu, J.; Yin, T.; Lu, X.J. T cell dysfunction in cancer immunity and immunotherapy. Front. Immunol. 2019, 10, 1719. [Google Scholar] [CrossRef] [PubMed]
  106. Xu, J.; Yu, S.; Wang, X.; Qian, Y.; Wu, W.; Zhang, S.; Zheng, B.; Wei, G.; Gao, S.; Cao, Z.; et al. High affinity of chlorin e6 to immunoglobulin G for intraoperative fluorescence image-guided cancer photodynamic and checkpoint blockade therapy. ACS Nano 2019, 13, 10242–10260. [Google Scholar] [CrossRef]
  107. Guo, Y.; Liu, Y.; Wu, W.; Ling, D.; Zhang, Q.; Zhao, P.; Hu, X. Indoleamine 2,3-dioxygenase (IDO) inhibitors and their nanomedicines for cancer immunotherapy. Biomaterials 2021, 276, 121018. [Google Scholar] [CrossRef] [PubMed]
  108. Wu, M.; Huang, Q.; Xie, Y.; Wu, X.; Ma, H.; Zhang, Y.; Xia, Y. Improvement of the anticancer efficacy of PD-1/PD-L1 blockade via combination therapy and PD-L1 regulation. J. Hematol. Oncol. 2022, 15, 24. [Google Scholar] [CrossRef]
  109. Pardoll, D.M. The blockade of immune checkpoints in cancer immunotherapy. Nat. Rev. Cancer 2012, 12, 252–264. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  110. Eggermont, A.M.M.; Blank, C.U.; Mandala, M.; Long, G.V.; Atkinson, V.; Dalle, S.; Haydon, A.; Lichinitser, M.; Khattak, A.; Carlino, M.S.; et al. Adjuvant pembrolizumab versus placebo in resected stage III melanoma. N. Engl. J. Med. 2018, 378, 1789–1801. [Google Scholar] [CrossRef] [PubMed]
  111. Wang, D.; Lin, J.; Yang, X.; Long, J.; Bai, Y.; Yang, X.; Mao, Y.; Sang, X.; Seery, S.; Zhao, H. Combination regimens with PD-1/PD-L1 immune checkpoint inhibitors for gastrointestinal malignancies. J. Hematol. Oncol. 2019, 12, 42. [Google Scholar] [CrossRef]
  112. Lommatzsch, M.; Bratke, K.; Stoll, P. Neoadjuvant PD-1 blockade in resectable lung cancer. N. Engl. J. Med. 2018, 379, e14. [Google Scholar] [PubMed]
  113. Madsen, S.J.; Christie, C.; Huynh, K.; Peng, Q.; Uzal, F.A.; Krasieva, T.B.; Hirschberg, H. Limiting glioma development by photodynamic therapy-generated macrophage vaccine and allo-stimulation: An in vivo histological study in rats. J. Biomed. Opt. 2018, 2, 028001. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  114. Etminan, N.; Peters, C.; Lakbir, D.; Bünemann, E.; Börger, V.; Sabel, M.C.; Hänggi, D.; Steiger, H.J.; Stummer, W.; Sorg, R.V. Heat-shock protein 70-dependent dendritic cell activation by 5-aminolevulinic acid-mediated photodynamic treatment of human glioblastoma spheroids in vitro. Br. J. Cancer 2011, 105, 961–969. [Google Scholar] [CrossRef] [PubMed]
  115. Shixiang, Y.; Xi, S.; Junliang, L.; Shanyi, Z.; Xingke, X.; Meiguang, Z.; Kai, W.; Fangcheng, L. Antitumor efficacy of a photodynamic therapy-generated dendritic cell glioma vaccine. Med. Oncol. 2011, 28, S453–S461. [Google Scholar] [CrossRef] [PubMed]
  116. Garg, A.D.; Vandenberk, L.; Koks, C.; Verschuere, T.; Boon, L.; Van Gool, S.W.; Agostinis, P. Dendritic cell vaccines based on immunogenic cell death elicit danger signals and T cell-driven rejection of high-grade glioma. Sci. Transl. Med. 2016, 8, 328ra27. [Google Scholar] [CrossRef] [PubMed]
  117. Bartusik-Aebisher, D.; Żołyniak, A.; Barnaś, E.; Machorowska-Pieniążek, A.; Oleś, P.; Kawczyk-Krupka, A.; Aebisher, D. The use of photodynamic therapy in the treatment of brain tumors—A review of the literature. Molecules 2022, 27, 6847. [Google Scholar] [CrossRef] [PubMed]
  118. Gunaydin, G.; Gedik, M.; Ayan, S. Photodynamic Therapy—Current Limitations and Novel Approaches. Front. Chem. 2021, 9, 691697. [Google Scholar] [CrossRef]
  119. Chen, R.; Aghi, M.K. Atypical meningiomas. Handb. Clin. Neurol. 2020, 170, 233–244. [Google Scholar] [PubMed]
  120. Kiesel, B.; Freund, J.; Reichert, D.; Wadiura, L.; Erkkilae, M.T.; Woehrer, A.; Hervey-Jumper, S.; Berger, M.S.; Widhalm, G. 5-ALA in suspected low-grade gliomas: Current Role, limitations, and new approaches. Front. Oncol. 2021, 11, 699301. [Google Scholar] [CrossRef] [PubMed]
  121. Kim, M.M.; Darafsheh, A. Light Sources and Dosimetry Techniques for Photodynamic Therapy. Photochem. Photobiol. 2020, 96, 280–294. [Google Scholar] [CrossRef] [Green Version]
  122. Salehpour, F.; Cassano, P.; Rouhi, N.; Hamblin, M.R.; De Taboada, L.; Farajdokht, F.; Mahmoudi, J. Penetration profiles of visible and near-infrared lasers and light-emitting diode light through the head tissues in animal and human species: A review of literature. Photobiomodul. Photomed. Laser Surg. 2019, 37, 581–595. [Google Scholar] [CrossRef] [PubMed]
  123. Genina, E.A.; Bashkatov, A.N.; Tuchina, D.K.; Dyachenko-Timoshina, P.A.; Navolokin, N.; Shirokov, A.; Khorovodov, A.; Terskov, A.; Klimova, M.; Mamedova, A.; et al. Optical properties of brain tissues at the different stages of glioma development in rats: Pilot study. Biomed. Opt. Express 2019, 10, 5182–5197. [Google Scholar] [CrossRef] [PubMed]
  124. Semyachkina-Glushkovskaya, O.; Abdurashitov, A.; Dubrovsky, A.; Klimova, M.; Agranovich, I.; Terskov, A.; Shirokov, A.; Vinnik, V.; Kuzmina, A.; Lezhnev, N.; et al. Photobiomodulation of lymphatic drainage and clearance: Perspective strategy for augmentation of meningeal lymphatic functions. Biomed. Opt. Express 2020, 11, 725–734. [Google Scholar] [CrossRef]
  125. Semyachkina-Glushkovskaya, O.; Fedosov, I.; Shirokov, A.; Vodovozova, E.; Alekseeva, A.; Khorovodov, A.; Blokhina, I.; Terskov, A.; Mamedova, A.; Klimova, M.; et al. Photomodulation of lymphatic delivery of liposomes to the brain bypassing the blood-brain barrier: New perspectives for glioma therapy. Nanophotonics 2021, 10, 3215–3227. [Google Scholar] [CrossRef]
  126. Semyachkina-Glushkovskaya, O.; Penzel, T.; Blokhina, I.; Khorovodov, A.; Fedosov, I.; Yu, T.; Karandin, G.; Evsukova, A.; Elovenko, D.; Adushkina, V.; et al. Night photostimulation of clearance of beta-amyloid from mouse brain: New strategies in preventing Alzheimer’s disease. Cells 2021, 10, 3289. [Google Scholar] [CrossRef] [PubMed]
  127. Zakharov, S.D.; Ivanov, A.V. Light-oxygen effect in cells and its potential applications in tumour therapy (review). Quantum Electron. 1999, 29, 1031–1053. [Google Scholar] [CrossRef]
  128. Park, J.H.; Lee, H.K. Current understanding of hypoxia in glioblastoma multiforme and its response to immunotherapy. Cancers 2022, 14, 1176. [Google Scholar] [CrossRef] [PubMed]
  129. Xiao, Z.; Halls, S.; Dickey, D.; Tulip, J.; Moore, R.B. Fractionated versus Standard Continuous Light Delivery in Interstitial Photodynamic Therapy of dunning Prostate Carcinomas. Clin. Cancer Res. 2007, 13, 7496–7505. [Google Scholar] [CrossRef] [Green Version]
  130. Maragkos, G.A.; Schüpper, A.J.; Lakomkin, N.; Sideras, P.; Price, G.; Baron, R.; Hamilton, T.; Haider, S.; Lee, I.Y.; Hadjipanayis, C.G.; et al. Fluorescence-guided high-grade glioma surgery more than four hours after 5-aminolevulinic acid administration. Front. Neurol. 2021, 12, 644804. [Google Scholar] [CrossRef]
  131. Stummer, W.; Pichlmeier, U.; Meinel, T.; Wiestler, O.D.; Zanella, F.; Reulen, H.J. Fluorescence-guided surgery with 5-aminolevulinic acid for resection of malignant glioma: A randomised controlled multicentre phase III trial. Lancet Oncol. 2006, 7, 392–401. [Google Scholar] [CrossRef]
  132. Senders, J.T.; Muskens, I.S.; Schnoor, R.; Karhade, A.V.; Cote, D.J.; Smith, T.R. Agents for fluorescence-guided glioma surgery: A systematic review of preclinical and clinical results. Acta Neurochir. 2017, 159, 151–167. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  133. Wan, S.; Parrish, J.A.; Anderson, R.R.; Madden, M. Transmittance of nonionizing radiation in human tissues. Photochem. Photobiol. 1981, 34, 679–681. [Google Scholar] [CrossRef] [PubMed]
  134. Lychagov, V.V.; Tuchin, V.V.; Vilensky, M.A.; Reznik, B.N.; Ichim, T.; De Taboada, L. Experimental study of NIR transmittance of the human skull. In Complex Dynamics and Fluctuations in Biomedical Photonics, 3rd ed.; International Society for Optics and Photonics: Bellingham, WA, USA, 2006; p. 60850T. [Google Scholar]
  135. Svaasand, L.O.; Ellingsen, R. Optical properties of human brain. Photochem. Photobiol. 1983, 38, 293–299. [Google Scholar] [CrossRef] [PubMed]
  136. Stolik, S.; Delgado, J.; Perez, A.; Anasagasti, L. Measurement of the penetration depths of red and near infrared light in human “ex vivo” tissues. J. Photochem. Photobiol. B 2000, 57, 90–93. [Google Scholar] [CrossRef] [PubMed]
  137. Jagdeo, J.R.; Adams, L.E.; Brody, N.I.; Siegel, D.M. Transcranial red and near infrared light transmission in a cadaveric model. PLoS ONE 2012, 7, e47460. [Google Scholar] [CrossRef]
  138. Yue, L.; Monge, M.; Ozgur, M.H. Simulation and measurement of transcranial near infrared light penetration. In Optical Interactions with Tissue and Cells, 26th ed.; International Society for Optics and Photonics: Bellingham, WA, USA, 2015; p. 93210S. [Google Scholar]
  139. Barrett, D.; Gonzalez-Lima, F. Transcranial infrared laser stimulation produces beneficial cognitive and emotional effects in humans. Neuroscience 2013, 230, 13–23. [Google Scholar] [CrossRef]
Figure 1. Schematic illustration of intracavitary PDT and interstitial PDT for GBM with an energy diagram of the oxygen response after PS excitation by an appropriate light wavelength. In the singlet state, energy is converted to heat by internal conversion or emitted as fluorescence. Then, PS in the excited singlet state can convert to the excited triplet state. In the triplet state, the energy can undergo a Type 1 or Type 2 redox reaction with the generation of the reactive oxygen species (ROS), which induces the death of the GBM cells via necrosis and/or apoptosis. This can also destruct the GBM vasculature and produce an acute inflammatory response that attracts leukocyte activation.
Figure 1. Schematic illustration of intracavitary PDT and interstitial PDT for GBM with an energy diagram of the oxygen response after PS excitation by an appropriate light wavelength. In the singlet state, energy is converted to heat by internal conversion or emitted as fluorescence. Then, PS in the excited singlet state can convert to the excited triplet state. In the triplet state, the energy can undergo a Type 1 or Type 2 redox reaction with the generation of the reactive oxygen species (ROS), which induces the death of the GBM cells via necrosis and/or apoptosis. This can also destruct the GBM vasculature and produce an acute inflammatory response that attracts leukocyte activation.
Pharmaceutics 14 02612 g001
Figure 2. The lymphatic clearance of tracer from the brain after crossing the OBBB. The figure illustrates the fluorescent microscopy of the clearance of TRITC-dextran 70 kDa (red color) from the brain via the meningeal lymphatics (green color, labelled by the specific antibodies LYVE-1 conjugated with Alexa 488). The intravenously injected TRITC-dextran is immediately observed in the Sagittal sinus (the main cerebral vein), and 30 min later, it is also present in the meningeal lymphatics due to the PDT-OBBB-mediated activation of the lymphatic clearance of tracer from the brain [43].
Figure 2. The lymphatic clearance of tracer from the brain after crossing the OBBB. The figure illustrates the fluorescent microscopy of the clearance of TRITC-dextran 70 kDa (red color) from the brain via the meningeal lymphatics (green color, labelled by the specific antibodies LYVE-1 conjugated with Alexa 488). The intravenously injected TRITC-dextran is immediately observed in the Sagittal sinus (the main cerebral vein), and 30 min later, it is also present in the meningeal lymphatics due to the PDT-OBBB-mediated activation of the lymphatic clearance of tracer from the brain [43].
Pharmaceutics 14 02612 g002
Figure 3. Schematic illustration of the depth of the penetration of light into the head and brain tissues, depending on the wavelength.
Figure 3. Schematic illustration of the depth of the penetration of light into the head and brain tissues, depending on the wavelength.
Pharmaceutics 14 02612 g003
Figure 4. PDT-OBBB modulation of brain tumor immunity. The cancer immunity cycle in GBM involves the APC release and lymphatic traffic of APCs into the dCLNs (1), where they activate CD4+/CD8+-T cells (2). The T-cells migrate from the dcLNs to the GBM, extravasate into the tumor tissues via OBBB (3), and, finally, the T cells induce GBM cytotoxicity, releasing more GBM antigens and contributing to brain tumor immunity.
Figure 4. PDT-OBBB modulation of brain tumor immunity. The cancer immunity cycle in GBM involves the APC release and lymphatic traffic of APCs into the dCLNs (1), where they activate CD4+/CD8+-T cells (2). The T-cells migrate from the dcLNs to the GBM, extravasate into the tumor tissues via OBBB (3), and, finally, the T cells induce GBM cytotoxicity, releasing more GBM antigens and contributing to brain tumor immunity.
Pharmaceutics 14 02612 g004
Publisher’s Note: MDPI stays neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Share and Cite

MDPI and ACS Style

Semyachkina-Glushkovskaya, O.; Terskov, A.; Khorovodov, A.; Telnova, V.; Blokhina, I.; Saranceva, E.; Kurths, J. Photodynamic Opening of the Blood–Brain Barrier and the Meningeal Lymphatic System: The New Niche in Immunotherapy for Brain Tumors. Pharmaceutics 2022, 14, 2612. https://doi.org/10.3390/pharmaceutics14122612

AMA Style

Semyachkina-Glushkovskaya O, Terskov A, Khorovodov A, Telnova V, Blokhina I, Saranceva E, Kurths J. Photodynamic Opening of the Blood–Brain Barrier and the Meningeal Lymphatic System: The New Niche in Immunotherapy for Brain Tumors. Pharmaceutics. 2022; 14(12):2612. https://doi.org/10.3390/pharmaceutics14122612

Chicago/Turabian Style

Semyachkina-Glushkovskaya, Oxana, Andrey Terskov, Alexander Khorovodov, Valeria Telnova, Inna Blokhina, Elena Saranceva, and Jürgen Kurths. 2022. "Photodynamic Opening of the Blood–Brain Barrier and the Meningeal Lymphatic System: The New Niche in Immunotherapy for Brain Tumors" Pharmaceutics 14, no. 12: 2612. https://doi.org/10.3390/pharmaceutics14122612

Note that from the first issue of 2016, this journal uses article numbers instead of page numbers. See further details here.

Article Metrics

Back to TopTop