Next Article in Journal
Characterization of the Compounds Present in Bougainvillea x buttiana (var. Rose) with Healing Activity in a Murine Model
Previous Article in Journal
Exploring the Anticancer Properties of 1,2,3-Triazole-Substituted Andrographolide Derivatives
Previous Article in Special Issue
Diverse Roles of Antibodies in Antibody–Drug Conjugates
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Near-Infrared Photoimmunotherapy in Brain Tumors—An Unexplored Frontier

1
Department of Biochemistry, School of Life Dentistry at Niigata, The Nippon Dental University, Niigata 951-8580, Japan
2
Department of Neurosurgery, Brain Research Institute, Niigata University, Niigata 951-8585, Japan
3
Department of Brain Tumor Biology, Brain Research Institute, Niigata University, Niigata 951-8585, Japan
4
Near InfraRed Photo-ImmunoTherapy Research Institute, Kansai Medical University, Hirakata, Osaka 573-1010, Japan
5
Faculty of Engineering, Niigata University, Niigata 950-2181, Japan
6
Startup Incubation Center, Shimadzu Corporation, Kyoto 604-8511, Japan
7
Advanced Treatment of Neurological Diseases Branch, Brain Research Institute, Niigata University, Niigata 951-8585, Japan
*
Author to whom correspondence should be addressed.
Pharmaceuticals 2025, 18(5), 751; https://doi.org/10.3390/ph18050751
Submission received: 31 March 2025 / Revised: 16 April 2025 / Accepted: 13 May 2025 / Published: 19 May 2025
(This article belongs to the Special Issue Antibody-Based Imaging and Targeted Therapy in Cancer)

Abstract

:
Near-infrared photoimmunotherapy (NIR-PIT) is a promising cancer treatment that uses near-infrared light to activate a conjugate of a monoclonal antibody (mAb) and a photoactivatable silica phthalocyanine dye (IRDye700DX: IR700). Unlike conventional photodynamic therapy (PDT), NIR-PIT selectively destroys targeted tumor cells while preserving the surrounding normal tissue and providing superior tissue penetration. Recently, NIR-PIT has been approved for the treatment of unresectable recurrent head and neck cancers in Japan. It induces highly selective cancer cell death; therefore, it is expected to be a new curative treatment option for various cancers, including brain tumors. In this review, we compare the principles of NIR-PIT and PDT and discuss the potential applications of NIR-PIT for brain tumors. We selected targetable proteins across various types of brain tumors and devised a strategy to effectively pass the mAb–IR700 conjugate through the blood–brain barrier (BBB), which is a significant challenge for NIR-PIT in treating brain tumors. Innovative approaches for delivering the mAb–IR700 conjugate across the BBB include exosomes, nanoparticle-based systems, and cell-penetrating peptides. Small-molecule compounds, such as affibodies, are anticipated to rapidly accumulate in tumors within intracranial models, and our preliminary experiments demonstrated rapid uptake. NIR-PIT also induces immunogenic cell death and activates the anti-tumor immune response. Overall, NIR-PIT is a promising approach for treating brain tumors. It has the potential to overcome the limitations of conventional therapies and offers new hope to patients with brain tumors.

1. Introduction

In brain tumor treatment, balancing the demands of removing as much tumor tissue as possible while preserving normal tissue is challenging. Conventional treatments, such as surgery, radiotherapy, and chemotherapy, each play an important role but also face several limitations. Notably, brain tumors often recur despite the combination of these treatments because of the difficulty of completely eliminating microinvasion at the tumor margins [1,2]. Therefore, photodynamic therapy (PDT) has been used to remove residual cancer cells around the extraction cavity or in unsafe areas of malignant brain tumors [3]. However, photosensitizers used in PDT can accumulate in normal cells and lead to nonspecific phototoxicity, which is not always an acceptable side effect [4,5].
Near-infrared photoimmunotherapy (NIR-PIT) has recently gained attention as an innovative light-based, selective cancer therapy. NIR-PIT is not an improved version of PDT; it offers a fundamentally different approach in terms of its mechanism and clinical potential. NIR-PIT utilizes a conjugate combined with a photoactivated silica phthalocyanine dye (IRDye700DX: IR700) and a monoclonal antibody. It can selectively destroy only targeted tumor cells without damaging the surrounding normal tissue. More importantly, this therapy induces immunogenic cell death in tumor cells and activates an anti-tumor immune response [6].
In recent years, numerous therapies utilizing photosensitizers and activating light have been reported. In these reports, the terms phototherapy and photoimmunotherapy were often used interchangeably without proper distinction, regardless of their mechanisms of action. Consequently, there is no globally standardized definition, which can lead to misunderstanding. In this review, we define and describe NIR-PIT as a therapy that employs IR700 as a photosensitizer and 690 nm near-infrared light to activate the IR700 conjugate, which is currently being clinically applied in Japan.
The selectivity of NIR-PIT is particularly valuable in the brain. Brain tumors, especially glioblastoma, exhibit infiltrative growth patterns and often lack a clear boundary with the normal brain tissue. In such cases, NIR-PIT overcomes the limitations of conventional treatment, offering the potential to completely eliminate these “invisible” tumor cells, representing a new therapeutic strategy to reduce the risk of recurrence. In recent years, extensive molecular-targeting studies have demonstrated the versatility of NIR-PIT. However, there have been few reports of NIR-PIT in brain tumors.
In this review, we compare the principles of PDT and NIR-PIT and also comprehensively explore the application of NIR-PIT in brain tumor treatment. We focus on brain-tumor-specific challenges, including penetrating the blood–brain barrier, antibody delivery systems, and potential molecular protein targets in various brain tumor types for NIR-PIT. In addition, we present preliminary data demonstrating the efficacy of NIR-PIT in brain tumor models. Furthermore, we discuss the anticipated immune responses and adverse effects associated with NIR-PIT in the brain. This review aimed to assess the potential of NIR-PIT as an unexplored frontier in the treatment of brain tumors.

2. PDT vs. NIR-PIT

Photodynamic therapy (PDT) originated in 1993, when T. Dougherty et al. from Canada performed PDT to prevent the recurrence of bladder cancer [7]. Targeted treatments have also been conducted for lung cancer and esophageal cancer; however, the photo-sensitizer, porfimer sodium (Photofrin®, absorption wavelength 630 nm), demonstrated poor selective accumulation in tumor cells and caused significant skin photo-sensitivity after administration [8]. In contrast, talaporfin sodium (Laserphyrin®, absorption wavelength 664 nm), approved for use in 2003 in Japan, is a photosensitizer that addresses the limitations of porfimer sodium [9].
The efficacy of talaporfin sodium in combination with a semiconductive laser emitting light against gliomas has been shown in a series of preclinical studies in Japan [10,11,12]. Subsequently, a two-center, single-arm phase II study testing the efficacy of talaporfin sodium in malignant gliomas was performed, with a 1-year overall survival of 95.5%, a 6-month local control rate of 91%, and, importantly, a relatively safe adverse event profile [13,14]. This study led to the healthcare approval of talaporfin sodium in combination with a semiconductive laser in 2013 in Japan for primary malignant brain tumors. The effectiveness of talaporfin sodium has subsequently been validated in newly diagnosed [14], as well as recurrent [15] glioblastoma at a single high-volume center in Japan.
Talaporfin sodium is intravenously administered 22–26 h before surgery [16]. It accumulates in significantly higher concentrations in cancer cells than in normal cells. The reason for this biodistribution may be the tendency of talaporfin sodium to preferentially combine with low-density lipoprotein (LDL). LDL supplies tissues with the cholesterol necessary to create membranes during cell division. Rapidly dividing cancer cells show increased uptake of LDL, which act as a “transporter” of the photosensitizer to the cancerous tissues [17].
When talaporfin sodium is irradiated with light (664 nm), the energy directs a photosensitizer molecule to the excited triplet state T1 [18]. A hydrogen or electron is transferred, which leads to the formation of free radicals and anion radicals of the photosensitizer and substrate. This process produces reactive oxygen species (ROS) inside the cells, leading to oxidative stress and resulting in the destruction of cancer cells [19,20].
Despite the similarities between PDT and NIR-PIT as cancer therapies that employ photosensitizers and light to activate photochemical reactions, their mechanisms are significantly different.
NIR-PIT utilizes a conjugate synthesized through chemical conjugation of IRDye700DX (IR700) N-hydroxysuccinimide (NHS) ester, a silicon phthalocyanine derivative, to monoclonal antibodies (mAbs) that target the receptors on cancer cells. Approximately three IR700 molecules are bound to each mAb molecule in this conjugate. When the conjugate binds to the receptor, near-infrared light (690 nm) is applied to induce photochemical ligand reactions within the conjugate. Subsequently, the conjugate releases hydrophilic side chains and becomes hydrophobic. This transformation leads the remaining molecules to aggregate and damage the cancer cell membrane, resulting in cell death due to osmotic pressure (Figure 1).
In IR700, the radical anion generated by the electron donor and near-infrared light is protonated by the oxonium ion to form a radical intermediate, which then elongates the Si-O bond and coordinates with a hydroxide ion, cleaving the axial ligand. Because phthalocyanine has a planar structure rich in π electrons, when the axial ligand is cleaved, the molecules stack due to π–π interactions, forming aggregates in aqueous solutions [21,22].
In NIR-PIT, internalization of the conjugate is not necessary to induce cell death. The importance of light irradiation being performed when the IR700 conjugate binds to the receptor on the cell surface should be stressed. Although some conjugates are internalized and generate ROS when exposed to near-infrared light, this is not the primary reason for cell damage [21,23,24].
NIR-PIT demonstrates remarkable selectivity for malignant cell elimination while preserving normal tissues. In Japan, the first NIR-PIT for human use was approved by the Pharmaceuticals and Medical Devices Agency (PMDA) in September 2020. In the U.S., NIR-PIT is currently being evaluated in a phase III clinical trial (ClinicalTrials.gov, NCT identifier: NCT03769506, study number: ASP-1929-301). Clinical validation targeting the epidermal growth factor receptor (EGFR) is particularly notable in head and neck squamous cell carcinoma (HNSCC) [25].
The differences between PDT and NIR-PIT are summarized in Table 1. A critical advantage of NIR-PIT is its significantly higher efficiency, owing to the optimal absorption characteristics of IR700. Talaporfin sodium used in PDT has a very intense absorption peak (Soret band) near 400 nm, a wavelength range not utilized in PDT because of poor tissue penetration, while IR700 has its maximum absorption at 690 nm, which is ideal for deep tissue penetration [26,27]. Generally, the 600–900 nm wavelength region is known as the “optical window”, allowing incident light between these wavelengths to penetrate more deeply into the tissue. This is the reason both NIR-PIT and PDT utilize light within this wavelength range [28]. Although talaporfin sodium exhibits a Q-band between 600 nm and 800 nm, its absorption efficiency is substantially lower than that of IR700 in this therapeutic window. This optimal absorption profile of IR700 at 690 nm allows NIR-PIT to achieve superior tumor-selective targeting, deeper tissue penetration, and enhanced therapeutic potency compared with PDT. Given these differences in photo-physical properties and mechanisms, NIR-PIT should not be viewed merely as a substitute for PDT in brain tumor treatment but rather as a revolutionary therapeutic approach with unprecedented possibilities.

3. Therapeutic Potential of NIR-PIT: Consideration of Targetable Surface Antigens Based on Brain Tumor Type

3.1. Glioblastoma

The incidence of glioblastoma varies from 3.19 to 4.17 per 100,000 persons-years [31,32], accounting for 14.5% of all central nervous system tumors and 48.6% of malignant brain tumors [33]. Prognosis is still dismal, with a median overall survival of 14.6 to 20.5 months, even after multimodal treatment, including surgery, radiation, and temozolomide [34,35]. In 2013, in Japan, intraoperative photodynamic therapy, involving presurgical intravenous injection of talaporfin sodium in combination with semiconductive laser emitting light, was approved. Intraoperative NIR-PIT is theoretically possible using a similar system, changing the wavelength of the light.
Several studies have been conducted looking at specific cell surface markers in glioblastomas. Chen et al. used the TCGA GBM database to find the top 10 expression-encoded cell surface proteins in glioblastomas, including CD44, CD68, podoplanin, CD163, and TREM2 [36]. Dutiot et al. studied the glioblastoma peptidome to uncover 10 tumor-associated antigens, such as BCAN, FABP7, and NRCAM, to be used in peptide vaccines [37].
Podoplanin is a type-1 transmembrane protein whose function is to bind to C-type lectin-like receptor-2 (CLEC-2) on platelets, inducing platelet aggregation. Physiologically, it is expressed on the lymphatic endothelium [38] with almost no expression in the normal brain or blood vessels [39,40]. We and others have previously shown that podoplanin is highly expressed in IDH-wildtype glioblastomas [41,42,43], whereas in IDH-mutant gliomas, its expression is suppressed by promoter methylation [44]. A preclinical study showed the effectiveness of chimeric antigen receptor (CAR)-T cell therapy targeting podoplanin in an orthotopic glioblastoma model. Podoplanin is also expressed in many tumors, including pleural mesotheliomas [45], lymphangioma [46], meningiomas [47], oral [48] and lung squamous cell carcinomas [49], and head and neck cancers [50], making it an exciting target for NIR-PIT. Preclinical studies showed the effectiveness of the podoplanin antibodies and IR700 conjugate in treating pleural mesothelioma [51] and oral cancer [52]. Kato et al. have produced many podoplanin antibodies, including cancer-specific antibodies that bind to post-translational glycans [53,54,55]. The same group has also developed humanized podoplanin antibodies [56,57].
Epidermal growth factor variant III (EGFRvIII) is an extensively studied surface marker of glioblastoma. It is a product of the EGRF gene with an in-frame deletion of exons 2–7 [58,59,60]. It is a tumor-specific cell surface antigen that constitutively activates the STAT [61] and PI3K–Akt pathways [62] and promotes angiogenesis and tumor growth [63,64,65]. It is known to be expressed mainly in the core of the tumor [66]. EGFRvIII is expressed in 25% of glioblastomas that are IDH-wildtype and EGFR amplified [67]. The peptide vaccine rindopepimut, which targets EGFRvIII, showed promising effects in a phase 2 clinical trial targeting glioblastomas [68], although a subsequent phase 3 trial combining rindopepimut with temozolomide in newly diagnosed glioblastomas expressing EGFRvIII failed to display effects in overall survival [69]. Preclinical studies have shown cytotoxicity of NIR-PIT using the EGFR antibody (ZEGFR:03115) in a subcutaneous U87-MGvIII xenograft model [70,71].
Cell adhesion molecule L1 (L1CAM) is a cell surface glycoprotein that promotes tumor migration in proliferation in glioblastomas [72]. L1CAM was found to be expressed in about 20% of both low- and high-grade gliomas [73]. L1CAM is also known to be expressed in supratentorial ependymomas harboring ZFTA fusions [74] and is used as a surrogate marker for this fusion. Other potential targets include GD2 for diffuse midline gliomas [75,76], EphA2 [77], TROP2 [78], IL13Rα2 [79,80], and B7-H3 [81,82,83] for glioblastomas.

3.2. Glioma Stem Cells

The literature contains a vast amount of data studying cancer stem cells in glioblastomas. Targeting glioma stem cells is an attractive option, as stem cells are known to be treatment-resistant and have pluripotency and self-renewal properties [84,85,86,87,88,89]. Proposed surface markers of glioma stem cells include CD133 [90,91], CD44 [92], and aldehyde dehydrogenases, such as ALDH1A3 [93]. NIR-PIT studies successfully targeting CD133 [94,95] in glioblastomas have been reported. CD44 is a stem cell marker in mesenchymal-type glioblastomas, the most aggressive type of glioblastoma. CD44–IR700 has been studied in many cancers, such as pancreatic cancer [96,97], triple-negative breast cancer [98], and epithelioid sarcoma [99], but remains to be studied in glioblastomas. ALDH has not been targeted in NIR-PIT studies, but ALDH inhibitors and ALDH peptide-based dendritic cell therapy targeting cancer stem cells [100] have been explored.

3.3. Brain Metastasis

Approximately 20% of all cancer patients develop brain metastases and often face a poor prognosis [101]. Against this background, an additional treatment option is required, and NIR-PIT for brain metastases would be a suitable therapeutic approach for these patients.
Lung cancer, breast cancer, and melanoma are the most common primary tumors that metastasize to the brain, accounting for ≥50%, 15–25%, and 5–20% of all brain metastases, respectively [102,103]. Ferguson et al. investigated protein expression on brain metastases from the tumors above to identify potential therapeutic targets. Based on their results and considering the mechanism of NIR-PIT targeting specific proteins on the cancer cell membrane, the targetable proteins are EGFR (expression rate: 66.7%), cMET (31.8%), PDL-1 (22%), PGP (20.1%) for non-small cell lung cancer (NSCLC), HER2 (23.3%) and EGFR (18.8%) for breast cancer, and cMET (36.4%) for melanoma [104]. In particular, HER2 can be an ideal therapeutic target since several studies have shown that HER2 expression is upregulated in brain metastases compared to that in primary breast cancer [105,106]. While PGP is a theoretical target, NIR-PIT targeting PGP seems impractical because it may damage the normal BBB tissues that express PGP [107]. To the best of our knowledge, there are no in vivo NIR-PIT studies using hematogenous brain metastasis models yet. However, NIR-PIT targeting HER2, PD-L1, or PGP has been reported in various extracranial cancers [108,109,110].
Although it remains unclear whether conjugates can cross the BBB, multiple studies have successfully imaged or treated brain metastases using monoclonal antibodies targeting specific proteins [111,112,113]. These reports demonstrated that monoclonal antibodies can cross the blood–brain barrier (BBB) and accumulate in brain metastases, suggesting the feasibility of treating brain metastases using NIR-PIT.

3.4. Malignant Meningioma

Meningiomas arise from arachnoid cap cells, forming extra-axial tumors that compress the brain and can eventually invade it. They account for approximately 25–40% of all primary brain tumors [114]. They are generally benign tumors, largely belonging to World Health Organization (WHO) grade 1 tumors. However, recent studies have discovered new prognostic markers, including brain invasion and molecular markers, such as TERT promoter mutation and CDKN2A homozygous deletion [115,116,117]. Integrated diagnosis has raised the percentage of WHO grade 2/3 meningiomas to as high as 38% [115], much higher than that of the ~10% in the pre-molecular era. Notably, grade 3 malignant meningiomas have a poor prognosis, with a 5-year survival rate of approximately 66% and a 10-year survival rate of 14–24% [118,119,120]. Symptomatic or progressively enlarging meningiomas require treatment, with surgical resection being the standard approach. While complete resection, including the dura mater, offers the potential for a cure in benign cases, localization of meningiomas around critical neurovascular structures, such as the dural sinuses, important arteries, cranial nerves, and the skull base, can make curative resection of the tumor by surgery impossible. Additionally, radiotherapy is frequently employed for recurrent, unresectable, or high-grade meningiomas; however, its efficacy remains limited for large tumor volumes and highly aggressive tumors [121]. Residual tumor, even in WHO grade 1 meningiomas, can be problematic. Meningiomas can regrow after radiation treatment. Therefore, there is a need for novel treatments for meningiomas.
Besides podoplanin, somatostatin receptors, especially somatostatin receptor 2 (SSTR-2) and somatostatin receptor 5 (SSTR-5), are frequently expressed in meningiomas and are promising targets for malignant meningioma [122,123,124]. SSTR-2 has been reported to be expressed in 64–100% of meningiomas [122,123,124,125,126,127,128,129], making it an interesting target for NIR-PIT. Importantly, SSTR-2 expression in meningiomas can be pre-operatively assessed by positron emission tomography (PET) using radiolabeled SSTR-2 ligands, such as 68Ga-DOTATE [130] or 68Ga-DOTATOC [131]. Other somatostatin SSTR2-targeted therapies, including somatostatin analogs [132,133] and peptide receptor radionuclide therapy with 90Y- and 177Lu-DOTATOC [134], are also under investigation.

3.5. Functional Pituitary Neuroendocrine Tumor (PitNET)

Neuroendocrine neoplasms (NENs) represent a heterogeneous group of epithelial tumors that arise in various organs, including the gastrointestinal tract and pancreas. They are characterized by morphological and functional features of neuroendocrine differentiation, such as secretory granules, synaptic-like vesicles, and the ability to produce amines and/or peptide hormones [135]. NENs are broadly classified into well-differentiated neuroendocrine tumors (NETs) and poorly differentiated neuroendocrine carcinomas (NECs) [136]. Treatment strategies for NETs depend on the primary site, tumor grade, and disease stage and include surgical resection, somatostatin analogs (SSAs), chemotherapy, and peptide receptor radionuclide therapy (PRRT) [137]. NETs often overexpress specific membrane receptors, particularly SSTRs, enabling both diagnostic imaging and receptor-targeted therapies. PRRT, in particular, has demonstrated efficacy in patients with advanced, progressive, somatostatin-receptor-positive NETs [138]. The clinical success of these receptor-based approaches highlights the therapeutic potential of membrane receptor expression in the management of NETs.
In the 2022 WHO classification (WHO Classification of Endocrine and Neuroendocrine Tumors, 5th Edition), the former term “pituitary adenoma” was replaced with “pituitary neuroendocrine tumor (PitNET)” [139]. While most PitNETs are managed with surgery and/or medical therapy, some PitNETs show treatment resistance or recurrence [140]. Functional PitNETs, like other NETs, frequently overexpress membrane receptors, such as SSTR2, SSTR5, and dopamine receptors, and thus may respond to SSAs [141] and, in selected cases, to PRRT [142]. Given this receptor profile, NIR-PIT, which selectively induces cell death via receptor-targeted antibodies conjugated with photoactivatable dyes, may provide a novel therapeutic option not only for malignant NETs but also for PitNETs with receptor overexpression. However, Volante et al. reported that in poorly differentiated NECs, expression of both SSTR2 and SSTR5 is reduced [143]. Therefore, in cases where PitNET progresses to high-grade pituitary neuroendocrine carcinoma (PitNEC), the effectiveness of SSTR-targeted NIR-PIT may be limited. Careful consideration is warranted in such cases, and the identification of alternative membrane targets may be necessary to develop effective therapies.

4. Designing the Optimal Partner for IR700 Conjugate

4.1. Challenges in Developing Ideal Conjugates for NIR-PIT in Brain Tumor

The BBB is a highly selective, semipermeable border consisting of brain microvascular endothelial cells connected by tight junctions and covered by pericytes and glial cells. It regulates the passage of molecules between the bloodstream and the brain microenvironment, preventing toxins and pathogens while allowing essential nutrients to pass through [144]. There are several transport mechanisms for substances crossing the BBB, including paracellular pathways, receptor-mediated transport, and cell-penetrating peptides. Generally, only certain gas molecules or highly lipid-soluble, positively charged molecules with a low molecular weight of less than 400 to 500 Da can enter the brain through paracellular pathways [145,146,147]. Approximately 98% of small molecules and nearly all large therapeutic molecules, such as monoclonal antibodies, cannot cross this barrier [146,148].
In primary brain tumors and brain metastases, the BBB is altered to the blood–brain–tumor barrier (BBTB), which is more permeable than the BBB [149]. However, the BBTB is highly heterogeneous, including non-uniform permeability and active efflux of molecules. Tellingen et al. reported that in low-grade gliomas, the BBTB is similar in structure and function to the normal BBB, whereas in high-grade gliomas, increased permeability could be identified in the BBTB. Furthermore, BBTB permeability is high in the bulk of the tumor but low or non-existent in the periphery in glioblastomas [150]. These factors result in inconsistent drug penetration and insufficient therapeutic accumulation in brain tumors [151,152].
Despite these challenging conditions, several antibody therapies for brain tumors, such as bevacizumab and rituximab, are already in clinical use [153]. Theoretically, rituximab, which targets CD20 on the surface of B cells, can be a candidate for the conjugate for NIR-PIT in primary central nervous system lymphoma. Nagaya et al. reported that NIR-PIT using the anti-CD20 rituximab–IR700 conjugate worked highly effectively for B-cell lymphoma [154]. However, studies have shown that a 0.1% to 4.4% concentration of rituximab was detected in cerebrospinal fluid compared with serum [155]. The range of concentration differences is likely due to heterogeneity in the BBTB of each patient. To demonstrate fully effective NIR-PIT, sufficient accumulation of the conjugate in the tumor is necessary. Therefore, even if rituximab stably binds to IR700, it may not be the ideal partner in some cases.
To perform NIR-PIT for brain tumors, the conjugates require properties that overcome these obstacles and effectively cross the BBB or BBTB, enabling sufficient accumulation in tumors. Therefore, various strategies for drug delivery have been explored [156,157].

4.2. The Epitope of Antibody for NIR-PIT Is in the Extracellular Domain

The location of an appropriate antibody is critical for the successful application of NIR-PIT. The antibodies used in NIR-PIT are conjugated to a photoactivatable dye, such as IR700 [6]; this section discusses NIR-PIT antibodies from the perspective of antibody-based therapies.
Among the five classes of immunoglobulin (Ig), IgG is predominantly used for therapeutic purposes in immunotherapy, including NIR-PIT. Currently, IgG (cetuximab) is clinically used for NIR-PIT. IgG is a large protein complex composed of two 55 kDa-heavy chains (H chains) and two 25 kDa-light chains (L-chains), linked by disulfide bonds, forming a Y-shaped structure [158]. Because IgG cannot readily pass through intact cell membranes while retaining its native structure and function, therapeutic antibodies are typically directed against extracellular antigens, including cell surface proteins and soluble antigens in the bloodstream [159]. Therapeutically relevant surface antigens include glycoantigens, cell adhesion molecules, receptors, transporters, ion channels, and immune-related markers. In addition, neoantigens classified as tumor-associated antigens may also serve as viable targets in cancer immunotherapy [160].
The distinctive feature of NIR-PIT is that monoclonal antibodies must recognize epitopes that are accessible on the extracellular surface of the plasma membrane. This critical distinction is often overlooked during screening. Failure to select antibodies that bind to accessible regions may result in inadequate photosensitizer delivery and compromised therapeutic efficacy. In other words, as long as they recognize the extracellular domain, not only monoclonal antibodies but also proteins, such as nanobodies, minibodies, and diabodies, enable NIR-PIT therapy.

4.3. New Strategies to Deliver IR700 Conjugate to the Brain

To achieve NIR-PIT for brain tumors, new strategies need to be developed to transport sufficient amounts of conjugates while maintaining the ability to bind to targeted receptors on cancer cells. Many different brain delivery platforms for antibodies have been studied, including exosomes, nanoparticle-based systems, and cell-penetrating peptides [161,162].
Chu et al. developed an antibody delivery platform using exosomes that can cross the BBB and effectively deliver bevacizumab, which has been approved to treat glioblastoma. Their animal experiments have illustrated that the introduction of exosomes–bevacizumab into the brain can improve the degeneration of pathological tissues, increase the apoptosis of tumor cells, and significantly extend the survival time [163]. Furthermore, the “Trojan horse” method enables the passage of biomolecules, such as antibodies, through the BBB. Wen et al. developed a two-step Trojan horse to enhance the brain penetration of the monoclonal antibody rituximab. They nano-encapsulated rituximab within a polymer layer that improved BBB penetration, increasing the central nervous system concentrations of the antibody by approximately tenfold compared to naked rituximab [164]. As mentioned above, rituximab is insufficient for use as an NIR-PIT conjugate because of its poor BBB permeability; however, using this method, it may be possible to achieve higher penetration.
While only monoclonal antibodies currently fulfill the role of delivering IR700 in clinical use, recent studies have reported NIR-PIT using alternative carriers, such as peptides, Fabs, and minibodies [165,166,167,168]. Watanabe et al. developed small and bivalent antibody fragments, including the anti-prostate-specific membrane antigen (PSMA) diabody (Db) and minibody (Mb), and compared them to IgG for their effectiveness as photoimmunotherapeutic agents. NIR-PIT with the same molar concentrations of the PSMA-Db–IR700, PSMA-Mb–IR700, and PSMA-IgG–IR700 conjugates showed similar therapeutic effects in vitro and in vivo. They concluded that the use of the PSMA-Db–IR700 conjugate resulted in the shortest time interval between injection and NIR exposure without compromising the therapeutic effects of photoimmunotherapy [169].
In terms of brain tumors, Burley et al. investigated NIR-PIT using an EGFR-specific Affibody conjugated to IR700 for glioblastoma. The Affibody–IR700 conjugate showed specific uptake in vitro and enabled imaging of EGFR expression in the orthotopic brain tumor model. The in vivo study demonstrated the therapeutic efficacy of this conjugate in subcutaneous glioma xenografts [70]. Importantly, it has been reported that there is a significantly higher accumulation of Affibody EGFR–IR800CW than cetuximab–IR680RD in the boundaries of glioma tumors, even though cetuximab has approximately 30 times greater affinity for EGFR than Affibody molecules [70]. Taken together, small molecules, such as Affibody, have the potential to be optimal partners for NIR-PIT in brain tumors.

4.4. Preliminary Data Showing Rapid Uptake of Affibody Conjugate in an Intracranial Xenograft Model

Small molecules, such as Affibodies, may efficiently cross the partially disrupted BBB. In the present study, we investigated the time taken for the EGFR Affibody–IR700 conjugate to reach the tumor in an intracranial brain tumor xenograft model. We used a Luminous Quester NX (Shimadzu Corp., Kyoto, Japan) to conduct real-time fluorescence imaging. This in vivo imaging system does not require a dark room and enables simultaneous fluorescence monitoring.
We allowed the growth of RSV-M (EGFR-expressing mouse brain tumor cells) tumors for 2 weeks after cell injection into C3H/HeSlc mice. For real-time imaging, the EGFR Affibody–IR700 conjugate was injected into the tail vein of each mouse. Fluorescence signals were detected in the cranial region in 10 s, reaching plateau levels after 15 s, followed by signal attenuation after 18 s (Figure 2a, Supplementary Materials Video S1). One hour after injection of the EGFR Affibody–IR700 conjugate, the brains were extracted, and the fluorescence intensity was evaluated (Figure 2b). After fixation, the brains were sectioned coronally for histological examination. Subsequent hematoxylin and eosin (H&E) staining confirmed the presence of cancer cells in regions exhibiting strong IR700 accumulation (Figure 2c).
Small molecules, such as Affibodies, likely facilitate more rapid tumor accumulation than full antibodies. Current clinical NIR-PIT protocols require IR700 conjugate injection 24 h prior to near-infrared light irradiation. However, our findings suggest that NIR-PIT utilizing small molecular carriers, such as Affibodies, could enable more flexible therapeutic approaches, including intraoperative NIR-PIT administration during surgery.

5. Immune Response After NIR-PIT

5.1. Mechanisms of NIR-PIT-Induced Immunogenic Cell Death in Brain Tumors

Immunogenic cell death (ICD) is known to trigger an adaptive immune response against cell-related antigens after damage-associated molecular patterns (DAMPs) are released upon cell death [170]. Ogawa et al. reported that when the cell membrane is physically destroyed during NIR-PIT, dying cells release DAMPs, which lead to ICD and the subsequent activation of the antitumor immune system that can address metastatic lesions [171]. It is well recognized that ROS production is essential for the release of DAMPs that lead to ICD [170]. However, when NIR-PIT damages the cancer cell membrane in a unique manner, ROS are not required for the release of DAMPs.
Męczyńska et al. confirmed the release of ROS by NIR-PIT using HER2 Affibody and concluded that ROS production releases ICD [172]. In addition, Nakajima et al. reported that the endocytosed mAb–IR700 conjugate is transported to the lysosome, where ROS induces necrotic cell death after near-infrared light irradiation [173]. Taken together, these results suggest that NIR-PIT induces ICD through two mechanisms: DAMPs released via physical membrane destruction, and ROS generation by IR700 internalized in lysosomes.
Both Ogawa et al. and Męczyńska et al. confirmed key ICD biomarkers, including the relocation of calreticulin, Hsp70, and Hsp90 to the cell surface, along with the rapid release of immunogenic signals, such as ATP and HMGB1, after NIR-PIT. These events induce the maturation of dendritic cells. The matured dendritic cells subsequently lead to CD8+ T-cell activation, enhancing antitumor immunity.
In a glioblastoma mouse model, NIR-PIT activated both CD4+ and CD8+ T cells while significantly increasing the expression of IL-1β and IL-6 cytokines, which can enhance the tumor immune system [71,174]. This suggests that NIR-PIT in the brain causes similar responses to activate the immune system as NIR-PIT in extracranial cancer.
In the in vivo NIR-PIT experiments we conducted using immunodeficient mice (BALB/c-nu), we observed microglia migrating into the brain tumor following near-infrared light irradiation [175]. Although many aspects remain unknown, these findings suggest that NIR-PIT effectively stimulates the immune system in the brain. While NIR-PIT may not eliminate all cancer cells during light irradiation, the remaining cancer cells may be targeted by the activated immune response.

5.2. Unique Challenges and Potential of NIR-PIT for Brain Tumors

Conventional cancer immunotherapies activate T cells with cytokines, immune checkpoint inhibitors, such as anti-PD1/PDL1 antibodies, immune suppressive cells, and CART therapy [176,177,178]. These therapies often cause systemic side effects, such as off-target effects, like autoimmune diseases [179]. Immunotherapy for brain tumors is even more difficult because the immune system in the brain has not been fully elucidated.
NIR-PIT only affects the area irradiated with near-infrared light, which is why side effects are unlikely to appear systemically. If NIR-PIT for brain tumors can also activate the immune system in a tumor-specific and partial manner, NIR-PIT could be the most suitable immunotherapy for brain tumors.
Okada et al. reported that Treg-targeted NIR-PIT using anti-CD25 antibodies was effective and that selective removal of Tregs by NIR-PIT activated CD8+ T cells and NK cells, demonstrating an “abscopal” effect that affected distant tumors [180]. Brain Tregs infiltrate the brain during inflammation, such as cerebral infarction. Ito et al. reported that the removal of brain Tregs in cerebral infarction leads to the activation of astrocytes (astrogliosis) and worsening of neurological symptoms. They also found that brain Tregs have characteristic gene expression that is not present in Tregs from other tissues and that brain Tregs interact with astrocytes and contribute to repairing brain damage [181].
Brain immunity has not yet been fully elucidated, and the details of the immune response to NIR-PIT in brain tumors have not been thoroughly examined, even at the basic experimental level. Therefore, considering these differences in Tregs, it may be difficult to control Tregs in the same manner as in extracranial cancers.
Furthermore, recent research has revealed that immune cells, such as monocytes and B cells, are supplied directly to the brain and spinal cord from the bone marrow within the skull. It has also been shown that these cells, which usually remain in the meninges, may infiltrate the brain and spinal cord parenchyma in response to inflammation or injury [182]. These cell infiltrations may occur after NIR-PIT of brain tumors. However, the details of the reaction remain unclear and need to be thoroughly investigated.

6. Treating Adverse Events in NIR-PIT for Brain Tumors

When NIR-PIT is applied to HNSCC patients in clinical practice, severe swelling is known to occur as a side effect [25,183]. Therefore, a tracheostomy is performed in patients with a risk of airway obstruction after NIR-PIT for HNSCC during the surgery. The area of swelling is not limited to the tumor site, and although it has been reported that COX2 inhibitors suppress swelling in mouse models after NIR-PIT, the underlying mechanism remains unclear [184]. Experiments conducted in mouse models have demonstrated that NIR-PIT in brain tumors causes swelling [71]. Consequently, the most important aspect of safety in NIR-PIT treatment is the control of transient swelling. This is critical when considering NIR-PIT treatment for brain tumors, as elevation of intracranial pressure can cause herniation. Performing a craniectomy first and then a cranioplasty after the swelling has subsided is possible, although it would involve multiple surgeries. Hinge/floating craniotomy [185] or expansive cranioplasty [186] are reasonable alternatives to decompressive craniectomy, which would mitigate the need for multiple surgeries. Osmotic agents, such as glycerol and mannitol, are regularly used to relieve intracranial pressure [187]. The usage of corticosteroids would also be an effective treatment, although their use may reduce secondary effects brought about by the immune response after NIR-PIT. Finally, bevacizumab, a vascular endothelial growth factor (VEGF)-A monoclonal antibody that produces potent anti-angiogenetic effects while relieving brain edema, is approved for use in glioblastomas. It is known to have corticosteroid-sparing effects [188]. Rigorous monitoring of brain edema after NIR-PIT will be mandatory, and the exact methods to control intracranial pressure remain to be elucidated.

7. NIR-PIT for Brain Tumors

As previously mentioned, applying NIR-PIT for brain tumors requires establishing a precise method to control intracranial pressure. If this can be achieved, it may allow for NIR-PIT of brain tumors, similar to the current NIT-PIT for head and neck tumors in Japan. For shallow lesions of tumors, a frontal diffuser is used to directly irradiate light forward, whereas for deep lesions of tumors, a cylindrical diffuser is inserted into the tumor and irradiates light in a cylindrical shape [189,190].
Moreover, given the unique environment of the brain, there may be unique methods to activate conjugates. For instance, employing longer-wavelength near-infrared light to penetrate deeper areas, minimizing scattered light with optical filters to enhance treatment efficiency, using pulse irradiation to activate photosensitizers more effectively, and possibly combining photosensitizers with X-ray-sensitive materials to improve access to deep tumors using X-rays may all be valid methods.
Currently, the most feasible and realistic approach is to use NIR-PIT for margins after tumor removal rather than attempting to treat the entire mass of the brain tumor. To perform NIR-PIT for brain tumors, it is possible to apply the techniques used in PDT, which are currently used in clinical practice.
NIR-PIT demonstrates greater selectivity than conventional treatments and is expected to become a promising new therapeutic approach for brain tumors. This technique may address the challenge of treating brain tumors: it aims to remove as much tumor tissue as possible while minimizing damage to the surrounding normal tissue.

8. Conclusions

NIR-PIT represents a revolutionary approach for brain tumor treatment through antibody-guided targeting combined with localized light activation. Although significant challenges remain, including optimal antibody delivery across the BBB, management of edema, and understanding brain-specific immune responses, the potential benefits are substantial.
NIR-PIT shows new therapeutic possibilities and illuminates a path toward an unexplored frontier, offering renewed hope for patients with brain tumors.

Supplementary Materials

The following supporting information can be downloaded at: https://www.mdpi.com/article/10.3390/ph18050751/s1, Video S1: Video showing rapid uptake of Affibody conjugate in an intracranial xenograft brain metastasis model.

Author Contributions

Conceptualization, H.Y. and M.N.; investigation, H.Y.; writing—original draft preparation, H.Y. and M.N.; writing—review and editing, all authors; supervision, M.O. (Makoto Oishi); funding acquisition, H.Y., S.S., T.T., R.O. and M.N. All authors have read and agreed to the published version of the manuscript.

Funding

This study was supported in part by the Japan Society for Promotion of Science KAKENHI, grant nos. 21K17102, 20K23067 (H.Y.), 24K12283 (S.S.), 25K12335 (T.T.), 22K16652 (R.O.), and 24K12238 (M.N.). M.N. has received research support from the New Sustainable Growth (NSG) group.

Institutional Review Board Statement

Although this paper is a review article, the relevant data shown in the present paper were obtained from experiments performed after the approval of the Institutional Review Board of Niigata University (approval #2022-0271). The animal experimental protocol for the preliminary experimental data in Section 4.3 was approved by the Laboratory Animal Investigation Committee of the School of Life Dentistry, Nippon Dental University, School of Dentistry (no. 232, date of approval: 31 May 2021).

Data Availability Statement

Not applicable.

Acknowledgments

The authors would like to acknowledge Takeyoshi Eda, Akiko Yoshii, and Tamami Murakami for their technical support and Hideyuki Sakata and Akihiro Ishikawa for their input on related subjects.

Conflicts of Interest

A.I. and H.S. are employees of Shimadzu Corporation. The other authors declare no conflicts of interest. Shimadzu Corporation had no role in the design of the study, the writing of the manuscript, or the decision to publish the results.

Abbreviations

The following abbreviations are used in this manuscript:
ALDHAldehyde dehydrogenases
BBBBlood–brain barrier
BBTBBlood–brain–tumor barrier
CAR-TChimeric antigen receptor-T
CLEC-2C-type lectin-like receptor-2
DAMPsDamage-associated molecular patterns
DBDiabody
EGFRvIIIEpidermal growth factor variant III
H&EHematoxylin and eosin
HNSCCHead and neck squamous cell carcinoma
ICDImmunogenic cell death
LDLLow-density lipoproteins
MBMinibody
NECNeuroendocrine carcinoma
NENNeuroendocrine neoplasm
NETNeuroendocrine tumor
NIR-PITNear-infrared photoimmunotherapy
NSCLCNon-small cell lung cancer
PDTPhotodynamic therapy
PETPositron emission tomography
Pit-NECPituitary neuroendocrine carcinoma
Pit-NETPituitary neuroendocrine tumor
PMDAPharmaceuticals and Medical Devices Agency
PRRTPeptide receptor radionuclide therapy
PSMAProstate-specific membrane antigen
ROSReactive oxygen species
SSASomatostatin analogue
SSTR-2Somatostatin receptor type 2
SSTR-5Somatostatin receptor type 5
VEGFVascular endothelial growth factor
WHOWorld Health Organization

References

  1. Fukuya, Y.; Ikuta, S.; Maruyama, T.; Nitta, M.; Saito, T.; Tsuzuki, S.; Chernov, M.; Kawamata, T.; Muragaki, Y. Tumor recurrence patterns after surgical resection of intracranial low-grade gliomas. J. Neuro-Oncol. 2019, 144, 519–528. [Google Scholar] [CrossRef] [PubMed]
  2. Tan, A.C.; Ashley, D.M.; López, 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]
  3. Cramer, S.W.; Chen, C.C. Photodynamic Therapy for the Treatment of Glioblastoma. Front. Surg. 2019, 6, 81. [Google Scholar] [CrossRef] [PubMed]
  4. 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]
  5. Allison, R.R.; Moghissi, K. Photodynamic Therapy (PDT): PDT Mechanisms. Clin. Endosc. 2013, 46, 24–29. [Google Scholar] [CrossRef]
  6. Mitsunaga, M.; Ogawa, M.; Kosaka, N.; Rosenblum, L.T.; Choyke, P.L.; Kobayashi, H. Cancer cell-selective in vivo near infrared photoimmunotherapy targeting specific membrane molecules. Nat. Med. 2011, 17, 1685–1691. [Google Scholar] [CrossRef]
  7. Dougherty, T.J. Photodynamic therapy. Photochem. Photobiol. 1993, 58, 895–900. [Google Scholar] [CrossRef]
  8. Nwogu, C.; Kloc, A.; Attwood, K.; Bshara, W.; Durrani, F.; Pandey, R. Porfimer Sodium Versus PS785 for Photodynamic Therapy (PDT) of Lung Cancer Xenografts in Mice. J. Surg. Res. 2021, 263, 245–250. [Google Scholar] [CrossRef] [PubMed]
  9. Yoshida, T.; Tokashiki, R.; Ito, H.; Shimizu, A.; Nakamura, K.; Hiramatsu, H.; Tsukahara, K.; Shimizu, S.; Takata, D.; Okamoto, I.; et al. Therapeutic effects of a new photosensitizer for photodynamic therapy of early head and neck cancer in relation to tissue concentration. Auris Nasus Larynx 2008, 35, 545–551. [Google Scholar] [CrossRef]
  10. Matsumura, H.; Akimoto, J.; Haraoka, J.; Aizawa, K. Uptake and retention of the photosensitizer mono-L-asparthyl chlorine e6 in experimental malignant glioma. Lasers Med. Sci. 2008, 23, 237–245. [Google Scholar] [CrossRef]
  11. Miki, Y.; Akimoto, J.; Yokoyama, S.; Homma, T.; Tsutsumi, M.; Haraoka, J.; Hirano, K.; Beppu, M. Photodynamic therapy in combination with talaporfin sodium induces mitochondrial apoptotic cell death accompanied with necrosis in glioma cells. Biol. Pharm. Bull. 2013, 36, 215–221. [Google Scholar] [CrossRef]
  12. Namatame, H.; Akimoto, J.; Matsumura, H.; Haraoka, J.; Aizawa, K. Photodynamic therapy of C6-implanted glioma cells in the rat brain employing second-generation photosensitizer talaporfin sodium. Photodiagn. Photodyn. Ther. 2008, 5, 198–209. [Google Scholar] [CrossRef] [PubMed]
  13. 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. JNS 2013, 119, 845–852. [Google Scholar] [CrossRef]
  14. Nitta, M.; Muragaki, Y.; Maruyama, T.; Iseki, H.; Komori, T.; Ikuta, S.; Saito, T.; Yasuda, T.; Hosono, J.; Okamoto, S.; et al. Role of photodynamic therapy using talaporfin sodium and a semiconductor laser in patients with newly diagnosed glioblastoma. J. Neurosurg. 2019, 131, 1361–1368. [Google Scholar] [CrossRef]
  15. Kobayashi, T.; Nitta, M.; Shimizu, K.; Saito, T.; Tsuzuki, S.; Fukui, A.; Koriyama, S.; Kuwano, A.; Komori, T.; Masui, K.; et al. Therapeutic Options for Recurrent Glioblastoma—Efficacy of Talaporfin Sodium Mediated Photodynamic Therapy. Pharmaceutics 2022, 14, 353. [Google Scholar] [CrossRef] [PubMed]
  16. Shimizu, K.; Nitta, M.; Komori, T.; Maruyama, T.; Yasuda, T.; Fujii, Y.; Masamune, K.; Kawamata, T.; Maehara, T.; Muragaki, Y. Intraoperative Photodynamic Diagnosis Using Talaporfin Sodium Simultaneously Applied for Photodynamic Therapy against Malignant Glioma: A Prospective Clinical Study. Front. Neurol. 2018, 9, 24. [Google Scholar] [CrossRef] [PubMed]
  17. Cruz, P.M.; Mo, H.; McConathy, W.J.; Sabnis, N.; Lacko, A.G. The role of cholesterol metabolism and cholesterol transport in carcinogenesis: A review of scientific findings, relevant to future cancer therapeutics. Front. Pharmacol. 2013, 4, 119. [Google Scholar] [CrossRef]
  18. Robertson, C.A.; Evans, D.H.; Abrahamse, H. Photodynamic therapy (PDT): A short review on cellular mechanisms and cancer research applications for PDT. J. Photochem. Photobiol. B 2009, 96, 1–8. [Google Scholar] [CrossRef]
  19. Ibarra, L.E.; Vilchez, M.L.; Caverzán, M.D.; Milla Sanabria, L.N. Understanding the glioblastoma tumor biology to optimize photodynamic therapy: From molecular to cellular events. J. Neurosci. Res. 2021, 99, 1024–1047. [Google Scholar] [CrossRef]
  20. Nowak-Stepniowska, A.; Pergoł, P.; Padzik-Graczyk, A. Photodynamic method of cancer diagnosis and therapy--mechanisms and applications. Postepy Biochem. 2013, 59, 53–63. [Google Scholar]
  21. Sato, K.; Ando, K.; Okuyama, S.; Moriguchi, S.; Ogura, T.; Totoki, S.; Hanaoka, H.; Nagaya, T.; Kokawa, R.; Takakura, H.; et al. Photoinduced Ligand Release from a Silicon Phthalocyanine Dye Conjugated with Monoclonal Antibodies: A Mechanism of Cancer Cell Cytotoxicity after Near-Infrared Photoimmunotherapy. ACS Cent. Sci. 2018, 4, 1559–1569. [Google Scholar] [CrossRef] [PubMed]
  22. Kobayashi, M.; Harada, M.; Takakura, H.; Ando, K.; Goto, Y.; Tsuneda, T.; Ogawa, M.; Taketsugu, T. Theoretical and Experimental Studies on the Near-Infrared Photoreaction Mechanism of a Silicon Phthalocyanine Photoimmunotherapy Dye: Photoinduced Hydrolysis by Radical Anion Generation. Chempluschem 2020, 85, 1959–1963. [Google Scholar] [CrossRef] [PubMed]
  23. Kobayashi, H.; Griffiths, G.L.; Choyke, P.L. Near-Infrared Photoimmunotherapy: Photoactivatable Antibody-Drug Conjugates (ADCs). Bioconjug. Chem. 2020, 31, 28–36. [Google Scholar] [CrossRef] [PubMed]
  24. Maruoka, Y.; Wakiyama, H.; Choyke, P.L.; Kobayashi, H. Near infrared photoimmunotherapy for cancers: A translational perspective. EBioMedicine 2021, 70, 103501. [Google Scholar] [CrossRef] [PubMed]
  25. Miyazaki, N.L.; Furusawa, A.; Choyke, P.L.; Kobayashi, H. Review of RM-1929 Near-Infrared Photoimmunotherapy Clinical Efficacy for Unresectable and/or Recurrent Head and Neck Squamous Cell Carcinoma. Cancers 2023, 15, 5117. [Google Scholar] [CrossRef]
  26. Akbar, A.; Khan, S.; Chatterjee, T.; Ghosh, M. Unleashing the power of porphyrin photosensitizers: Illuminating breakthroughs in photodynamic therapy. J. Photochem. Photobiol. B 2023, 248, 112796. [Google Scholar] [CrossRef]
  27. Inagaki, F.F.; Fujimura, D.; Furusawa, A.; Okada, R.; Wakiyama, H.; Kato, T.; Choyke, P.L.; Kobayashi, H. Fluorescence Imaging of Tumor-Accumulating Antibody-IR700 Conjugates Prior to Near-Infrared Photoimmunotherapy (NIR-PIT) Using a Commercially Available Camera Designed for Indocyanine Green. Mol. Pharm. 2021, 18, 1238–1246. [Google Scholar] [CrossRef]
  28. Mallidi, S.; Anbil, S.; Bulin, A.L.; Obaid, G.; Ichikawa, M.; Hasan, T. Beyond the Barriers of Light Penetration: Strategies, Perspectives and Possibilities for Photodynamic Therapy. Theranostics 2016, 6, 2458–2487. [Google Scholar] [CrossRef]
  29. Li, B.; Lin, L. Internal light source for deep photodynamic therapy. Light. Sci. Appl. 2022, 11, 85. [Google Scholar] [CrossRef]
  30. Kiesslich, T.; Neureiter, D.; Alinger, B.; Jansky, G.L.; Berlanda, J.; Mkrtchyan, V.; Ocker, M.; Plaetzer, K.; Berr, F. Uptake and phototoxicity of meso-tetrahydroxyphenyl chlorine are highly variable in human biliary tract cancer cell lines and correlate with markers of differentiation and proliferation. Photochem. Photobiol. Sci. 2010, 9, 734–743. [Google Scholar] [CrossRef]
  31. Batash, R.; Asna, N.; Schaffer, P.; Francis, N.; Schaffer, M. Glioblastoma Multiforme, Diagnosis and Treatment; Recent Literature Review. Curr. Med. Chem. 2017, 24, 3002–3009. [Google Scholar] [CrossRef] [PubMed]
  32. Fabbro-Peray, P.; Zouaoui, S.; Darlix, A.; Fabbro, M.; Pallud, J.; Rigau, V.; Mathieu-Daude, H.; Bessaoud, F.; Bauchet, F.; Riondel, A.; et al. Association of patterns of care, prognostic factors, and use of radiotherapy-temozolomide therapy with survival in patients with newly diagnosed glioblastoma: A French national population-based study. J. Neuro-Oncol. 2019, 142, 91–101. [Google Scholar] [CrossRef] [PubMed]
  33. Grochans, S.; Cybulska, A.M.; Simińska, D.; Korbecki, J.; Kojder, K.; Chlubek, D.; Baranowska-Bosiacka, I. Epidemiology of Glioblastoma Multiforme-Literature Review. Cancers 2022, 14, 2412. [Google Scholar] [CrossRef]
  34. Stupp, R.; Mason, W.P.; van den Bent, M.J.; Weller, M.; Fisher, B.; Taphoorn, M.J.B.; Belanger, K.; Brandes, A.A.; Marosi, C.; Bogdahn, U.; et al. Radiotherapy plus concomitant and adjuvant temozolomide for glioblastoma. N. Engl. J. Med. 2005, 352, 987–996. [Google Scholar] [CrossRef] [PubMed]
  35. Stupp, R.; Taillibert, S.; Kanner, A.A.; Kesari, S.; Steinberg, D.M.; Toms, S.A.; Taylor, L.P.; Lieberman, F.; Silvani, A.; Fink, K.L.; et al. Maintenance therapy with tumor-treating fields plus temozolomide vs temozolomide alone for glioblastoma: A randomized clinical trial. JAMA 2015, 314, 2535–2543. [Google Scholar] [CrossRef]
  36. Chen, T.; Chen, J.; Zhu, Y.; Li, Y.; Wang, Y.; Chen, H.; Wang, J.; Li, X.; Liu, Y.; Li, B.; et al. CD163, a novel therapeutic target, regulates the proliferation and stemness of glioma cells via casein kinase 2. Oncogene 2019, 38, 1183–1199. [Google Scholar] [CrossRef]
  37. Dutoit, V.; Herold-Mende, C.; Hilf, N.; Schoor, O.; Beckhove, P.; Bucher, J.; Dorsch, K.; Flohr, S.; Fritsche, J.; Lewandrowski, P.; et al. Exploiting the glioblastoma peptidome to discover novel tumour-associated antigens for immunotherapy. Brain 2012, 135, 1042–1054. [Google Scholar] [CrossRef]
  38. Breiteneder-Geleff, S.; Soleiman, A.; Kowalski, H.; Horvat, R.; Amann, G.; Kriehuber, E.; Diem, K.; Weninger, W.; Tschachler, E.; Alitalo, K.; et al. Angiosarcomas express mixed endothelial phenotypes of blood and lymphatic capillaries: Podoplanin as a specific marker for lymphatic endothelium. Am. J. Pathol. 1999, 154, 385–394. [Google Scholar] [CrossRef]
  39. Wicki, A.; Christofori, G. The potential role of podoplanin in tumour invasion. Br. J. Cancer 2007, 96, 1–5. [Google Scholar] [CrossRef]
  40. Riedl, J.; Preusser, M.; Nazari, P.M.; Posch, F.; Panzer, S.; Marosi, C.; Birner, P.; Thaler, J.; Brostjan, C.; Lötsch, D.; et al. Podoplanin expression in primary brain tumors induces platelet aggregation and increases risk of venous thromboembolism. Blood 2017, 129, 1831–1839. [Google Scholar] [CrossRef]
  41. Watanabe, J.; Natsumeda, M.; Okada, M.; Kanemaru, Y.; Tsukamoto, Y.; Oishi, M.; Kakita, A.; Fujii, Y. Podoplanin Expression and IDH-Wildtype Status Predict Venous Thromboembolism in Patients with High-Grade Gliomas in the Early Postoperative Period. World Neurosurg. 2019, 128, e982–e988. [Google Scholar] [CrossRef] [PubMed]
  42. Mir Seyed Nazari, P.; Riedl, J.; Preusser, M.; Posch, F.; Thaler, J.; Marosi, C.; Birner, P.; Ricken, G.; Hainfellner, J.A.; Pabinger, I.; et al. Combination of isocitrate dehydrogenase 1 (IDH1) mutation and podoplanin expression in brain tumors identifies patients at high or low risk of venous thromboembolism. J. Thromb. Haemost. 2018, 16, 1121–1127. [Google Scholar] [CrossRef] [PubMed]
  43. Ando, K.; Natsumeda, M.; Kawamura, M.; Shirakawa, K.; Okada, M.; Tsukamoto, Y.; Eda, T.; Watanabe, J.; Saito, S.; Takahashi, H.; et al. Elevated ratio of C-type lectin-like receptor 2 level and platelet count (C2PAC) aids in the diagnosis of post-operative venous thromboembolism in IDH-wildtype gliomas. Thromb. Res. 2023, 223, 36–43. [Google Scholar] [CrossRef] [PubMed]
  44. Sun, C.; Xiao, L.; Zhao, Y.; Shi, J.; Yuan, Y.; Gu, Y.; Zhang, F.; Gao, X.; Yang, Y.; Yang, R.; et al. Wild-Type IDH1 and Mutant IDH1 Opposingly Regulate Podoplanin Expression in Glioma. Transl. Oncol. 2020, 13, 100758. [Google Scholar] [CrossRef]
  45. Abe, S.; Kaneko, M.K.; Tsuchihashi, Y.; Izumi, T.; Ogasawara, S.; Okada, N.; Sato, C.; Tobiume, M.; Otsuka, K.; Miyamoto, L.; et al. Antitumor effect of novel anti-podoplanin antibody NZ-12 against malignant pleural mesothelioma in an orthotopic xenograft model. Cancer Sci. 2016, 107, 1198–1205. [Google Scholar] [CrossRef]
  46. Ji, R.C.; Eshita, Y.; Xing, L.; Miura, M. Multiple expressions of lymphatic markers and morphological evolution of newly formed lymphatics in lymphangioma and lymph node lymphangiogenesis. Microvasc. Res. 2010, 80, 195–201. [Google Scholar] [CrossRef]
  47. Shibahara, J.; Kashima, T.; Kikuchi, Y.; Kunita, A.; Fukayama, M. Podoplanin is expressed in subsets of tumors of the central nervous system. Virchows Arch. 2006, 448, 493–499. [Google Scholar] [CrossRef]
  48. Inoue, H.; Tsuchiya, H.; Miyazaki, Y.; Kikuchi, K.; Ide, F.; Sakashita, H.; Kusama, K. Podoplanin expressing cancer-associated fibroblasts in oral cancer. Tumour Biol. 2014, 35, 11345–11352. [Google Scholar] [CrossRef]
  49. Yurugi, Y.; Wakahara, M.; Matsuoka, Y.; Sakabe, T.; Kubouchi, Y.; Haruki, T.; Nosaka, K.; Miwa, K.; Araki, K.; Taniguchi, Y.; et al. Podoplanin Expression in Cancer-associated Fibroblasts Predicts Poor Prognosis in Patients with Squamous Cell Carcinoma of the Lung. Anticancer. Res. 2017, 37, 207–213. [Google Scholar] [CrossRef]
  50. Ramos-Vega, V.; Venegas Rojas, B.; Donoso Torres, W. Immunohistochemical analysis of cancer-associated fibroblasts and podoplanin in head and neck cancer. Med. Oral. Patol. Oral. Cir. Bucal 2020, 25, e268–e276. [Google Scholar] [CrossRef]
  51. Nishinaga, Y.; Sato, K.; Yasui, H.; Taki, S.; Takahashi, K.; Shimizu, M.; Endo, R.; Koike, C.; Kuramoto, N.; Nakamura, S.; et al. Targeted Phototherapy for Malignant Pleural Mesothelioma: Near-Infrared Photoimmunotherapy Targeting Podoplanin. Cells 2020, 9, 1019. [Google Scholar] [CrossRef] [PubMed]
  52. Kato, T.; Furusawa, A.; Okada, R.; Inagaki, F.; Wakiyama, H.; Furumoto, H.; Fukushima, H.; Okuyama, S.; Choyke, P.L.; Kobayashi, H. Near-Infrared Photoimmunotherapy Targeting Podoplanin-Expressing Cancer Cells and Cancer-Associated Fibroblasts. Mol. Cancer Ther. 2023, 22, 75–88. [Google Scholar] [CrossRef] [PubMed]
  53. Kato, Y.; Kaneko, M.K. A cancer-specific monoclonal antibody recognizes the aberrantly glycosylated podoplanin. Sci. Rep. 2014, 4, 5924. [Google Scholar] [CrossRef]
  54. Tanaka, T.; Suzuki, H.; Ohishi, T.; Kaneko, M.K.; Kato, Y. A Cancer-Specific Anti-Podoplanin Monoclonal Antibody, PMab-117-mG2a Exerts Antitumor Activities in Human Tumor Xenograft Models. Cells 2024, 13, 1833. [Google Scholar] [CrossRef] [PubMed]
  55. Yamada, S.; Ogasawara, S.; Kaneko, M.K.; Kato, Y. LpMab-23: A Cancer-Specific Monoclonal Antibody Against Human Podoplanin. Monoclon. Antib. Immunodiagn. Immunother. 2017, 36, 72–76. [Google Scholar] [CrossRef]
  56. Suzuki, H.; Ohishi, T.; Kaneko, M.K.; Kato, Y. A Humanized and Defucosylated Antibody against Podoplanin (humLpMab-23-f) Exerts Antitumor Activities in Human Lung Cancer and Glioblastoma Xenograft Models. Cancers 2023, 15, 5080. [Google Scholar] [CrossRef]
  57. Ishikawa, A.; Waseda, M.; Ishii, T.; Kaneko, M.K.; Kato, Y.; Kaneko, S. Improved anti-solid tumor response by humanized anti-podoplanin chimeric antigen receptor transduced human cytotoxic T cells in an animal model. Genes Cells 2022, 27, 549–558. [Google Scholar] [CrossRef]
  58. Wikstrand, C.J.; Hale, L.P.; Batra, S.K.; Hill, M.L.; Humphrey, P.A.; Kurpad, S.N.; McLendon, R.E.; Moscatello, D.; Pegram, C.N.; Reist, C.J.; et al. Monoclonal antibodies against EGFRvIII are tumor specific and react with breast and lung carcinomas and malignant gliomas. Cancer Res. 1995, 55, 3140–3148. [Google Scholar]
  59. Nishikawa, R.; Ji, X.D.; Harmon, R.C.; Lazar, C.S.; Gill, G.N.; Cavenee, W.K.; Huang, H.J. A mutant epidermal growth factor receptor common in human glioma confers enhanced tumorigenicity. Proc. Natl. Acad. Sci. USA 1994, 91, 7727–7731. [Google Scholar] [CrossRef]
  60. Gupta, P.; Han, S.Y.; Holgado-Madruga, M.; Mitra, S.S.; Li, G.; Nitta, R.T.; Wong, A.J. Development of an EGFRvIII specific recombinant antibody. BMC Biotechnol. 2010, 10, 72. [Google Scholar] [CrossRef]
  61. Fan, Q.W.; Cheng, C.K.; Gustafson, W.C.; Charron, E.; Zipper, P.; Wong, R.A.; Chen, J.; Lau, J.; Knobbe-Thomsen, C.; Weller, M.; et al. EGFR phosphorylates tumor-derived EGFRvIII driving STAT3/5 and progression in glioblastoma. Cancer Cell 2013, 24, 438–449. [Google Scholar] [CrossRef] [PubMed]
  62. Guo, G.; Gong, K.; Wohlfeld, B.; Hatanpaa, K.J.; Zhao, D.; Habib, A.A. Ligand-Independent EGFR Signaling. Cancer Res. 2015, 75, 3436–3441. [Google Scholar] [CrossRef] [PubMed]
  63. Katanasaka, Y.; Kodera, Y.; Kitamura, Y.; Morimoto, T.; Tamura, T.; Koizumi, F. Epidermal growth factor receptor variant type III markedly accelerates angiogenesis and tumor growth via inducing c-myc mediated angiopoietin-like 4 expression in malignant glioma. Mol. Cancer 2013, 12, 31. [Google Scholar] [CrossRef]
  64. Inda, M.M.; Bonavia, R.; Mukasa, A.; Narita, Y.; Sah, D.W.; Vandenberg, S.; Brennan, C.; Johns, T.G.; Bachoo, R.; Hadwiger, P.; et al. Tumor heterogeneity is an active process maintained by a mutant EGFR-induced cytokine circuit in glioblastoma. Genes Dev. 2010, 24, 1731–1745. [Google Scholar] [CrossRef] [PubMed]
  65. Zanca, C.; Villa, G.R.; Benitez, J.A.; Thorne, A.H.; Koga, T.; D’Antonio, M.; Ikegami, S.; Ma, J.; Boyer, A.D.; Banisadr, A.; et al. Glioblastoma cellular cross-talk converges on NF-κB to attenuate EGFR inhibitor sensitivity. Genes Dev. 2017, 31, 1212–1227. [Google Scholar] [CrossRef]
  66. Nozawa, T.; Okada, M.; Natsumeda, M.; Eda, T.; Abe, H.; Tsukamoto, Y.; Okamoto, K.; Oishi, M.; Takahashi, H.; Fujii, Y.; et al. EGFRvIII Is Expressed in Cellular Areas of Tumor in a Subset of Glioblastoma. Neurol. Med. Chir. 2019, 59, 89–97. [Google Scholar] [CrossRef]
  67. Maire, C.L.; Ligon, K.L. Molecular pathologic diagnosis of epidermal growth factor receptor. Neuro Oncol. 2014, 16 (Suppl. S8), viii1–viii6. [Google Scholar] [CrossRef]
  68. Reardon, D.A.; Desjardins, A.; Vredenburgh, J.J.; O’Rourke, D.M.; Tran, D.D.; Fink, K.L.; Nabors, L.B.; Li, G.; Bota, D.A.; Lukas, R.V.; et al. Rindopepimut with Bevacizumab for Patients with Relapsed EGFRvIII-Expressing Glioblastoma (ReACT): Results of a Double-Blind Randomized Phase II Trial. Clin. Cancer Res. 2020, 26, 1586–1594. [Google Scholar] [CrossRef]
  69. Weller, M.; Butowski, N.; Tran, D.D.; Recht, L.D.; Lim, M.; Hirte, H.; Ashby, L.; Mechtler, L.; Goldlust, S.A.; Iwamoto, F.; et al. Rindopepimut with temozolomide for patients with newly diagnosed, EGFRvIII-expressing glioblastoma (ACT IV): A randomised, double-blind, international phase 3 trial. Lancet Oncol. 2017, 18, 1373–1385. [Google Scholar] [CrossRef]
  70. Burley, T.A.; Mączyńska, J.; Shah, A.; Szopa, W.; Harrington, K.J.; Boult, J.K.R.; Mrozek-Wilczkiewicz, A.; Vinci, M.; Bamber, J.C.; Kaspera, W.; et al. Near-infrared photoimmunotherapy targeting EGFR-Shedding new light on glioblastoma treatment. Int. J. Cancer 2018, 142, 2363–2374. [Google Scholar] [CrossRef]
  71. Mączyńska, J.; Raes, F.; Da Pieve, C.; Turnock, S.; Boult, J.K.R.; Hoebart, J.; Niedbala, M.; Robinson, S.P.; Harrington, K.J.; Kaspera, W.; et al. Triggering anti-GBM immune response with EGFR-mediated photoimmunotherapy. BMC Med. 2022, 20, 16. [Google Scholar] [CrossRef]
  72. Anderson, H.J.; Galileo, D.S. Small-molecule inhibitors of FGFR, integrins and FAK selectively decrease L1CAM-stimulated glioblastoma cell motility and proliferation. Cell. Oncol. 2016, 39, 229–242. [Google Scholar] [CrossRef]
  73. Zeng, J.; Xi, S.-Y.; Wang, F.; Liao, H.-D.; Yang, Y.-Z.; Hu, W.-M. L1CAM High Expression Associates with Poor Prognosis in Glioma but Does Not Correlate with C11orf95-RELA Fusion. BioMed Res. Int. 2020, 2020, 1353284. [Google Scholar] [CrossRef] [PubMed]
  74. Malgulwar, P.B.; Nambirajan, A.; Pathak, P.; Faruq, M.; Rajeshwari, M.; Singh, M.; Suri, V.; Sarkar, C.; Sharma, M.C. C11orf95-RELA fusions and upregulated NF-KB signalling characterise a subset of aggressive supratentorial ependymomas that express L1CAM and nestin. J. Neuro-Oncol. 2018, 138, 29–39. [Google Scholar] [CrossRef]
  75. Majzner, R.G.; Mahdi, J.; Ramakrishna, S.; Patel, S.; Chinnasamy, H.; Yeom, K.; Schultz, L.; Barsan, V.; Richards, R.; Campen, C.; et al. Major tumor regression in H3K27M-mutated diffuse midline glioma (DMG) following sequential intravenous (IV) and intracerbroventricular (ICV) delivery of GD2-CAR T cells. Cancer Res. 2022, 24 (Suppl. S1), i20–i21. [Google Scholar]
  76. Majzner, R.G.; Ramakrishna, S.; Yeom, K.W.; Patel, S.; Chinnasamy, H.; Schultz, L.M.; Richards, R.M.; Jiang, L.; Barsan, V.; Mancusi, R.; et al. GD2-CAR T cell therapy for H3K27M-mutated diffuse midline gliomas. Nature 2022, 603, 934–941. [Google Scholar] [CrossRef] [PubMed]
  77. Hatano, M.; Eguchi, J.; Tatsumi, T.; Kuwashima, N.; Dusak, J.E.; Kinch, M.S.; Pollack, I.F.; Hamilton, R.L.; Storkus, W.J.; Okada, H. EphA2 as a glioma-associated antigen: A novel target for glioma vaccines. Neoplasia 2005, 7, 717–722. [Google Scholar] [CrossRef]
  78. Hou, J.; Lv, A.; Deng, Q.; Zhang, G.; Hu, X.; Cui, H. TROP2 promotes the proliferation and metastasis of glioblastoma cells by activating the JAK2/STAT3 signaling pathway. Oncol. Rep. 2019, 41, 753–764. [Google Scholar] [CrossRef]
  79. Park, D.H.; Bhojnagarwala, P.S.; Liaw, K.; Bordoloi, D.; Tursi, N.J.; Zhao, S.; Binder, Z.A.; O’Rourke, D.; Weiner, D.B. Novel tri-specific T-cell engager targeting IL-13Rα2 and EGFRvIII provides long-term survival in heterogeneous GBM challenge and promotes antitumor cytotoxicity with patient immune cells. J. Immunother. Cancer 2024, 12, e009604. [Google Scholar] [CrossRef]
  80. Newman, J.P.; Wang, G.Y.; Arima, K.; Guan, S.P.; Waters, M.R.; Cavenee, W.K.; Pan, E.; Aliwarga, E.; Chong, S.T.; Kok, C.Y.L.; et al. Interleukin-13 receptor alpha 2 cooperates with EGFRvIII signaling to promote glioblastoma multiforme. Nat. Commun. 2017, 8, 1913. [Google Scholar] [CrossRef]
  81. Tachi, T.; Kijima, N.; Kuroda, H.; Ikeda, S.; Murakami, K.; Nakagawa, T.; Yaga, M.; Nakagawa, K.; Utsugi, R.; Hirayama, R.; et al. Antitumor effects of intracranial injection of B7-H3-targeted Car-T and Car-Nk cells in a patient-derived glioblastoma xenograft model. Cancer Immunol. Immunother. 2024, 73, 256. [Google Scholar] [CrossRef]
  82. Inthanachai, T.; Boonkrai, C.; Phakham, T.; Pisitkun, T.; Thaiwong, R.; Chuthaphakdikun, V.; Sakunrangsit, N.; Limprasutr, V.; Chinsuwan, T.; Hirankarn, N.; et al. Novel B7-H3 CAR T cells show potent antitumor effects in glioblastoma: A preclinical study. J. Immunother. Cancer 2025, 13, e010083. [Google Scholar] [CrossRef] [PubMed]
  83. Zhang, C.; Zhang, Z.; Li, F.; Shen, Z.; Qiao, Y.; Li, L.; Liu, S.; Song, M.; Zhao, X.; Ren, F.; et al. Large-scale analysis reveals the specific clinical and immune features of B7-H3 in glioma. Oncoimmunology 2018, 7, e1461304. [Google Scholar] [CrossRef] [PubMed]
  84. Eyler, C.E.; Rich, J.N. Survival of the fittest: Cancer stem cells in therapeutic resistance and angiogenesis. J. Clin. Oncol. 2008, 26, 2839–2845. [Google Scholar] [CrossRef]
  85. Frank, N.Y.; Schatton, T.; Frank, M.H. The therapeutic promise of the cancer stem cell concept. J. Clin. Investig. 2010, 120, 41–50. [Google Scholar] [CrossRef] [PubMed]
  86. Visvader, J.E.; Lindeman, G.J. Cancer stem cells in solid tumours: Accumulating evidence and unresolved questions. Nat. Rev. Cancer 2008, 8, 755–768. [Google Scholar] [CrossRef]
  87. Wicha, M.S.; Liu, S.; Dontu, G. Cancer stem cells: An old idea--a paradigm shift. Cancer Res. 2006, 66, 1883–1890, discussion 1895–1896. [Google Scholar] [CrossRef]
  88. Dean, M.; Fojo, T.; Bates, S. Tumour stem cells and drug resistance. Nat. Rev. Cancer 2005, 5, 275–284. [Google Scholar] [CrossRef]
  89. Reya, T.; Morrison, S.J.; Clarke, M.F.; Weissman, I.L. Stem cells, cancer, and cancer stem cells. Nature 2001, 414, 105–111. [Google Scholar] [CrossRef]
  90. Kahlert, U.D.; Suwala, A.K.; Koch, K.; Natsumeda, M.; Orr, B.A.; Hayashi, M.; Maciaczyk, J.; Eberhart, C.G. Pharmacologic Wnt Inhibition Reduces Proliferation, Survival, and Clonogenicity of Glioblastoma Cells. J. Neuropathol. Exp. Neurol. 2015, 74, 889–900. [Google Scholar] [CrossRef]
  91. Joyce, T.; Jagasia, S.; Tasci, E.; Camphausen, K.; Krauze, A.V. An Overview of CD133 as a Functional Unit of Prognosis and Treatment Resistance in Glioblastoma. Curr. Oncol. 2023, 30, 8278–8293. [Google Scholar] [CrossRef]
  92. Behnan, J.; Isakson, P.; Joel, M.; Cilio, C.; Langmoen, I.A.; Vik-Mo, E.O.; Badn, W. Recruited brain tumor-derived mesenchymal stem cells contribute to brain tumor progression. Stem Cells 2014, 32, 1110–1123. [Google Scholar] [CrossRef]
  93. Mao, P.; Joshi, K.; Li, J.; Kim, S.H.; Li, P.; Santana-Santos, L.; Luthra, S.; Chandran, U.R.; Benos, P.V.; Smith, L.; et al. Mesenchymal glioma stem cells are maintained by activated glycolytic metabolism involving aldehyde dehydrogenase 1A3. Proc. Natl. Acad. Sci. USA 2013, 110, 8644–8649. [Google Scholar] [CrossRef]
  94. Jing, H.; Weidensteiner, C.; Reichardt, W.; Gaedicke, S.; Zhu, X.; Grosu, A.L.; Kobayashi, H.; Niedermann, G. Imaging and Selective Elimination of Glioblastoma Stem Cells with Theranostic Near-Infrared-Labeled CD133-Specific Antibodies. Theranostics 2016, 6, 862–874. [Google Scholar] [CrossRef] [PubMed]
  95. Sakuma, M.; Kita, S.; Higuchi, H. Quantitative evaluation of malignant gliomas damage induced by photoactivation of IR700 dye. Sci. Technol. Adv. Mater. 2016, 17, 473–482. [Google Scholar] [CrossRef] [PubMed]
  96. Wolf, I.; Schultze-Seemann, S.; Gratzke, C.; Wolf, P. Targeting CD44 and EpCAM with Antibody Dye Conjugates for the Photoimmunotherapy of Prostate Cancer. Antibodies 2025, 14, 5. [Google Scholar] [CrossRef] [PubMed]
  97. Yaku, H.; Takahashi, K.; Okada, H.; Kobiyama, K.; Shiokawa, M.; Uza, N.; Kodama, Y.; Ishii, K.J.; Seno, H. Near-infrared photoimmunotherapy as a complementary modality to in situ vaccine in a preclinical pancreatic cancer model. Biochem. Biophys. Res. Commun. 2024, 737, 150534. [Google Scholar] [CrossRef]
  98. Mungra, N.; Nsole Biteghe, F.A.; Huysamen, A.M.; Hardcastle, N.S.; Bunjun, R.; Naran, K.; Lang, D.; Richter, W.; Hunter, R.; Barth, S. An Investigation into the In Vitro Targeted Killing of CD44-Expressing Triple-Negative Breast Cancer Cells Using Recombinant Photoimmunotherapeutics Compared to Auristatin-F-Based Antibody-Drug Conjugates. Mol. Pharm. 2024, 21, 4098–4115. [Google Scholar] [CrossRef]
  99. Jin, J.; Barnett, J.D.; Mironchik, Y.; Gross, J.; Kobayashi, H.; Levin, A.; Bhujwalla, Z.M. Photoimmunotheranostics of epithelioid sarcoma by targeting CD44 or EGFR. Transl. Oncol. 2024, 45, 101966. [Google Scholar] [CrossRef]
  100. Liao, F.; Zhang, J.; Hu, Y.; Najafabadi, A.H.; Moon, J.J.; Wicha, M.S.; Kaspo, B.; Whitfield, J.; Chang, A.E.; Li, Q. Efficacy of an ALDH peptide-based dendritic cell vaccine targeting cancer stem cells. Cancer Immunol. Immunother. 2022, 71, 1959–1973. [Google Scholar] [CrossRef]
  101. Achrol, A.S.; Rennert, R.C.; Anders, C.; Soffietti, R.; Ahluwalia, M.S.; Nayak, L.; Peters, S.; Arvold, N.D.; Harsh, G.R.; Steeg, P.S.; et al. Brain metastases. Nat. Rev. Dis. Primers 2019, 5, 5. [Google Scholar] [CrossRef]
  102. Lah, T.T.; Novak, M.; Breznik, B. Brain malignancies: Glioblastoma and brain metastases. Semin. Cancer Biol. 2020, 60, 262–273. [Google Scholar] [CrossRef]
  103. Wilhelm, I.; Molnár, J.; Fazakas, C.; Haskó, J.; Krizbai, I.A. Role of the blood-brain barrier in the formation of brain metastases. Int. J. Mol. Sci. 2013, 14, 1383–1411. [Google Scholar] [CrossRef] [PubMed]
  104. Ferguson, S.D.; Zheng, S.; Xiu, J.; Zhou, S.; Khasraw, M.; Brastianos, P.K.; Kesari, S.; Hu, J.; Rudnick, J.; Salacz, M.E.; et al. Profiles of brain metastases: Prioritization of therapeutic targets. Int. J. Cancer 2018, 143, 3019–3026. [Google Scholar] [CrossRef]
  105. Palmieri, D.; Bronder, J.L.; Herring, J.M.; Yoneda, T.; Weil, R.J.; Stark, A.M.; Kurek, R.; Vega-Valle, E.; Feigenbaum, L.; Halverson, D.; et al. Her-2 overexpression increases the metastatic outgrowth of breast cancer cells in the brain. Cancer Res. 2007, 67, 4190–4198. [Google Scholar] [CrossRef] [PubMed]
  106. Mitra, D.; Clark, J.W.; Shih, H.A.; Oh, K.S.; Brastianos, P.K.; Wo, J.Y.; Strickland, M.R.; Curry, W.T.; Parikh, A.R.; Corcoran, R.B.; et al. Enrichment of HER2 Amplification in Brain Metastases from Primary Gastrointestinal Malignancies. Oncologist 2019, 24, 193–201. [Google Scholar] [CrossRef] [PubMed]
  107. Schinkel, A.H. P-Glycoprotein, a gatekeeper in the blood-brain barrier. Adv. Drug Deliv. Rev. 1999, 36, 179–194. [Google Scholar] [CrossRef]
  108. Nagaya, T.; Nakamura, Y.; Sato, K.; Harada, T.; Choyke, P.L.; Hodge, J.W.; Schlom, J.; Kobayashi, H. Near infrared photoimmunotherapy with avelumab, an anti-programmed death-ligand 1 (PD-L1) antibody. Oncotarget 2017, 8, 8807–8817. [Google Scholar] [CrossRef]
  109. Mao, C.; Qu, P.; Miley, M.J.; Zhao, Y.; Li, Z.; Ming, X. P-glycoprotein targeted photodynamic therapy of chemoresistant tumors using recombinant Fab fragment conjugates. Biomater. Sci. 2018, 6, 3063–3074. [Google Scholar] [CrossRef]
  110. Yamashita, S.; Kojima, M.; Onda, N.; Yoshida, T.; Shibutani, M. Trastuzumab-based near-infrared photoimmunotherapy in xenograft mouse of breast cancer. Cancer Med. 2023, 12, 4579–4589. [Google Scholar] [CrossRef]
  111. Kurihara, H.; Hamada, A.; Yoshida, M.; Shimma, S.; Hashimoto, J.; Yonemori, K.; Tani, H.; Miyakita, Y.; Kanayama, Y.; Wada, Y.; et al. (64)Cu-DOTA-trastuzumab PET imaging and HER2 specificity of brain metastases in HER2-positive breast cancer patients. EJNMMI Res. 2015, 5, 8. [Google Scholar] [CrossRef]
  112. Nienhuis, P.H.; Antunes, I.F.; Glaudemans, A.; Jalving, M.; Leung, D.; Noordzij, W.; Slart, R.; de Vries, E.F.J.; Hospers, G.A.P. (18)F-BMS986192 PET Imaging of PD-L1 in Metastatic Melanoma Patients with Brain Metastases Treated with Immune Checkpoint Inhibitors: A Pilot Study. J. Nucl. Med. 2022, 63, 899–905. [Google Scholar] [CrossRef] [PubMed]
  113. Mitsogianni, M.; Trontzas, I.P.; Gomatou, G.; Ioannou, S.; Syrigos, N.K.; Kotteas, E.A. The changing treatment of metastatic her2-positive breast cancer. Oncol. Lett. 2021, 21, 287. [Google Scholar] [CrossRef] [PubMed]
  114. Ostrom, Q.T.; Price, M.; Neff, C.; Cioffi, G.; Waite, K.A.; Kruchko, C.; Barnholtz-Sloan, J.S. CBTRUS Statistical Report: Primary Brain and Other Central Nervous System Tumors Diagnosed in the United States in 2016–2020. Neuro Oncol. 2023, 12 (Suppl. S2), iv1–iv99. [Google Scholar] [CrossRef] [PubMed]
  115. Driver, J.; Hoffman, S.E.; Tavakol, S.; Woodward, E.; Maury, E.A.; Bhave, V.; Greenwald, N.F.; Nassiri, F.; Aldape, K.; Zadeh, G.; et al. A molecularly integrated grade for meningioma. Neuro Oncol. 2021, 24, 796–808. [Google Scholar] [CrossRef]
  116. Louis, D.N. World Health Organization Classification of Tumours of the Central Nervous System, 5th ed.; International Agency for Research on Cancer: Lyon, France, 2021. [Google Scholar]
  117. Sahm, F.; Aldape, K.D.; Brastianos, P.K.; Brat, D.J.; Dahiya, S.; von Deimling, A.; Giannini, C.; Gilbert, M.R.; Louis, D.N.; Raleigh, D.R.; et al. cIMPACT-NOW update 8: Clarifications on molecular risk parameters and recommendations for WHO grading of meningiomas. Neuro Oncol. 2024, 27, 319–330. [Google Scholar] [CrossRef]
  118. Walsh, K.A.-O.; Price, M.; Neff, C.; Komisarow, J.M.; Wimberly, C.E.; Kruchko, C.A.-O.X.; Barnholtz-Sloan, J.S.; Ostrom, Q.T. The joint impacts of sex and race/ethnicity on incidence of grade 1 versus grades 2–3 meningioma across the lifespan. Neuro-Oncol. Adv. 2023, 5, i5–i12. [Google Scholar] [CrossRef]
  119. Maier, A.D.; Meddis, A.; Mirian, C.; Haslund-Vinding, J.; Bartek, J.; Krog, S.M.; Nguyen, T.U.P.; Areškevičiūtė, A.; Melchior, L.C.; Heegaard, S.; et al. Gene expression analysis during progression of malignant meningioma compared to benign meningioma. J. Neurosurg. 2022, 138, 1302–1312. [Google Scholar] [CrossRef]
  120. Tosefsky, K.; Rebchuk, A.D.; Wang, J.Z.; Ellenbogen, Y.; Drexler, R.; Ricklefs, F.L.; Sauvigny, T.; Schüller, U.; Cutler, C.B.; Lucke-Wold, B.; et al. Grade 3 meningioma survival and recurrence outcomes in an international multicenter cohort. J. Neurosurg. 2024, 140, 393–403. [Google Scholar] [CrossRef]
  121. Klungtvedt, V.; Aunan-Diop, J.S.; Poulsen, F.R.; Pedersen, C.B.; Wismann, J.; Wang, E.W.; Dahlrot, R.H.; Halle, B. Radiotherapy for World Health Organization Grade 1 and 2 Intracranial Meningiomas: A Retrospective Analysis of Efficacy. World Neurosurg. 2025, 197, 123858. [Google Scholar] [CrossRef]
  122. Silva, C.B.; Ongaratti, B.R.; Trott, G.; Haag, T.; Ferreira, N.P.; Leães, C.G.; Pereira-Lima, J.F.; Oliveira Mda, C. Expression of somatostatin receptors (SSTR1-SSTR5) in meningiomas and its clinicopathological significance. Int. J. Clin. Exp. Pathol. 2015, 8, 13185–13192. [Google Scholar] [PubMed]
  123. Dutour, A.; Kumar, U.; Panetta, R.; Ouafik, L.; Fina, F.; Sasi, R.; Patel, Y.C. Expression of somatostatin receptor subtypes in human brain tumors. Int. J. Cancer 1998, 76, 620–627. [Google Scholar] [CrossRef]
  124. Arena, S.; Barbieri, F.; Thellung, S.; Pirani, P.; Corsaro, A.; Villa, V.; Dadati, P.; Dorcaratto, A.; Lapertosa, G.; Ravetti, J.L.; et al. Expression of somatostatin receptor mRNA in human meningiomas and their implication in in vitro antiproliferative activity. J. Neuro-Oncol. 2004, 66, 155–166. [Google Scholar] [CrossRef]
  125. Schulz, S.; Pauli, S.U.; Schulz, S.; Händel, M.; Dietzmann, K.; Firsching, R.; Höllt, V. Immunohistochemical determination of five somatostatin receptors in meningioma reveals frequent overexpression of somatostatin receptor subtype sst2A. Clin. Cancer Res. 2000, 6, 1865–1874. [Google Scholar] [PubMed]
  126. Durand, A.; Champier, J.; Jouvet, A.; Labrousse, F.; Honnorat, J.; Guyotat, J.; Fèvre-Montange, M. Expression of c-Myc, neurofibromatosis Type 2, somatostatin receptor 2 and erb-B2 in human meningiomas: Relation to grades or histotypes. Clin. Neuropathol. 2008, 27, 334–345. [Google Scholar] [CrossRef] [PubMed]
  127. Barresi, V.; Alafaci, C.; Salpietro, F.; Tuccari, G. Sstr2A immunohistochemical expression in human meningiomas: Is there a correlation with the histological grade, proliferation or microvessel density? Oncol. Rep. 2008, 20, 485–492. [Google Scholar] [CrossRef]
  128. Agaimy, A.; Buslei, R.; Coras, R.; Rubin, B.P.; Mentzel, T. Comparative study of soft tissue perineurioma and meningioma using a five-marker immunohistochemical panel. Histopathology 2014, 65, 60–70. [Google Scholar] [CrossRef]
  129. Dijkstra, B.M.; Motekallemi, A.; den Dunnen, W.F.A.; Jeltema, J.R.; van Dam, G.M.; Kruyt, F.A.E.; Groen, R.J.M. SSTR-2 as a potential tumour-specific marker for fluorescence-guided meningioma surgery. Acta Neurochir. 2018, 160, 1539–1546. [Google Scholar] [CrossRef]
  130. Kunz, W.G.; Jungblut, L.M.; Kazmierczak, P.M.; Vettermann, F.J.; Bollenbacher, A.; Tonn, J.C.; Schichor, C.; Rominger, A.; Albert, N.L.; Bartenstein, P.; et al. Improved Detection of Transosseous Meningiomas Using (68)Ga-DOTATATE PET/CT Compared with Contrast-Enhanced MRI. J. Nucl. Med. 2017, 58, 1580–1587. [Google Scholar] [CrossRef]
  131. Afshar-Oromieh, A.; Giesel, F.L.; Linhart, H.G.; Haberkorn, U.; Haufe, S.; Combs, S.E.; Podlesek, D.; Eisenhut, M.; Kratochwil, C. Detection of cranial meningiomas: Comparison of ⁶⁸Ga-DOTATOC PET/CT and contrast-enhanced MRI. Eur. J. Nucl. Med. Mol. Imaging 2012, 39, 1409–1415. [Google Scholar] [CrossRef]
  132. Graillon, T.; Sanson, M.; Campello, C.; Idbaih, A.; Peyre, M.; Peyrière, H.; Basset, N.; Autran, D.; Roche, C.; Kalamarides, M.; et al. Everolimus and Octreotide for Patients with Recurrent Meningioma: Results from the Phase II CEVOREM Trial. Clin. Cancer Res. 2020, 26, 552–557. [Google Scholar] [CrossRef] [PubMed]
  133. Graillon, T.; Romano, D.; Defilles, C.; Saveanu, A.; Mohamed, A.; Figarella-Branger, D.; Roche, P.H.; Fuentes, S.; Chinot, O.; Dufour, H.; et al. Octreotide therapy in meningiomas: In vitro study, clinical correlation, and literature review. J. Neurosurg. 2017, 127, 660–669. [Google Scholar] [CrossRef] [PubMed]
  134. Marincek, N.; Radojewski, P.; Dumont, R.A.; Brunner, P.; Müller-Brand, J.; Maecke, H.R.; Briel, M.; Walter, M.A. Somatostatin receptor-targeted radiopeptide therapy with 90Y-DOTATOC and 177Lu-DOTATOC in progressive meningioma: Long-term results of a phase II clinical trial. J. Nucl. Med. 2015, 56, 171–176. [Google Scholar] [CrossRef]
  135. Hofland, J.; Kaltsas, G.; de Herder, W.W. Advances in the Diagnosis and Management of Well-Differentiated Neuroendocrine Neoplasms. Endocr. Rev. 2020, 41, 371–403. [Google Scholar] [CrossRef]
  136. Rindi, G.; Mete, O.; Uccella, S.; Basturk, O.; La Rosa, S.; Brosens, L.A.A.; Ezzat, S.; de Herder, W.W.; Klimstra, D.S.; Papotti, M.; et al. Overview of the 2022 WHO Classification of Neuroendocrine Neoplasms. Endocr. Pathol. 2022, 33, 115–154. [Google Scholar] [CrossRef]
  137. Modlin, I.M.; Oberg, K.; Chung, D.C.; Jensen, R.T.; de Herder, W.W.; Thakker, R.V.; Caplin, M.; Delle Fave, G.; Kaltsas, G.A.; Krenning, E.P.; et al. Gastroenteropancreatic neuroendocrine tumours. Lancet Oncol. 2008, 9, 61–72. [Google Scholar] [CrossRef]
  138. Strosberg, J.; El-Haddad, G.; Wolin, E.; Hendifar, A.; Yao, J.; Chasen, B.; Mittra, E.; Kunz, P.L.; Kulke, M.H.; Jacene, H.; et al. Phase 3 Trial of (177)Lu-Dotatate for Midgut Neuroendocrine Tumors. N. Engl. J. Med. 2017, 376, 125–135. [Google Scholar] [CrossRef]
  139. Tsukamoto, T.; Miki, Y. Imaging of pituitary tumors: An update with the 5th WHO Classifications-part 1. Pituitary neuroendocrine tumor (PitNET)/pituitary adenoma. Jpn. J. Radiol. 2023, 41, 789–806. [Google Scholar] [CrossRef] [PubMed]
  140. Chiloiro, S.; Costa, D.; Lauretta, R.; Mercuri, V.; Sbardella, E.; Samperi, I.; Appetecchia, M.; Bianchi, A.; Giampietro, A.; Gargiulo, P.; et al. Partial response to first generation SSA guides the choice and predict the outcome of second line therapy in acromegaly. Endocrine 2022, 78, 343–353. [Google Scholar] [CrossRef]
  141. Puig Domingo, M. Treatment of acromegaly in the era of personalized and predictive medicine. Clin. Endocrinol. 2015, 83, 3–14. [Google Scholar] [CrossRef]
  142. Marques, P. The Effects of Peptide Receptor Radionuclide Therapy on the Neoplastic and Normal Pituitary. Cancers 2023, 15, 2710. [Google Scholar] [CrossRef] [PubMed]
  143. Volante, M.; Brizzi, M.P.; Faggiano, A.; La Rosa, S.; Rapa, I.; Ferrero, A.; Mansueto, G.; Righi, L.; Garancini, S.; Capella, C.; et al. Somatostatin receptor type 2A immunohistochemistry in neuroendocrine tumors: A proposal of scoring system correlated with somatostatin receptor scintigraphy. Mod. Pathol. 2007, 20, 1172–1182. [Google Scholar] [CrossRef]
  144. Abbott, N.J.; Patabendige, A.A.; Dolman, D.E.; Yusof, S.R.; Begley, D.J. Structure and function of the blood-brain barrier. Neurobiol. Dis. 2010, 37, 13–25. [Google Scholar] [CrossRef] [PubMed]
  145. Wu, S.K.; Tsai, C.L.; Huang, Y.; Hynynen, K. Focused Ultrasound and Microbubbles-Mediated Drug Delivery to Brain Tumor. Pharmaceutics 2020, 13, 15. [Google Scholar] [CrossRef] [PubMed]
  146. Khawli, L.A.; Prabhu, S. Drug delivery across the blood-brain barrier. Mol. Pharm. 2013, 10, 1471–1472. [Google Scholar] [CrossRef]
  147. Patel, M.M.; Patel, B.M. Crossing the Blood-Brain Barrier: Recent Advances in Drug Delivery to the Brain. CNS Drugs 2017, 31, 109–133. [Google Scholar] [CrossRef]
  148. Nam, L.; Coll, C.; Erthal, L.C.S.; de la Torre, C.; Serrano, D.; Martínez-Máñez, R.; Santos-Martínez, M.J.; Ruiz-Hernández, E. Drug Delivery Nanosystems for the Localized Treatment of Glioblastoma Multiforme. Materials 2018, 11, 779. [Google Scholar] [CrossRef]
  149. Tiwary, S.; Morales, J.E.; Kwiatkowski, S.C.; Lang, F.F.; Rao, G.; McCarty, J.H. Metastatic Brain Tumors Disrupt the Blood-Brain Barrier and Alter Lipid Metabolism by Inhibiting Expression of the Endothelial Cell Fatty Acid Transporter Mfsd2a. Sci. Rep. 2018, 8, 8267. [Google Scholar] [CrossRef] [PubMed]
  150. van Tellingen, O.; Yetkin-Arik, B.; de Gooijer, M.C.; Wesseling, P.; Wurdinger, T.; de Vries, H.E. Overcoming the blood-brain tumor barrier for effective glioblastoma treatment. Drug Resist. Updates. 2015, 19, 1–12. [Google Scholar] [CrossRef]
  151. 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]
  152. Steeg, P.S. The blood-tumour barrier in cancer biology and therapy. Nat. Rev. Clin. Oncol. 2021, 18, 696–714. [Google Scholar] [CrossRef] [PubMed]
  153. Motomura, K.; Sasaki, K.; Sugii, N.; Yamaguchi, S.; Inoue, H.; Oshima, A.; Tanaka, K.; Otani, Y.; Shirahata, M.; Shibahara, I.; et al. Cost of medical care for malignant brain tumors at hospitals in the Japan Clinical Oncology Group brain-tumor study group. Jpn. J. Clin. Oncol. 2024, 54, 1123–1131. [Google Scholar] [CrossRef] [PubMed]
  154. Nagaya, T.; Nakamura, Y.; Sato, K.; Harada, T.; Choyke, P.L.; Kobayashi, H. Near infrared photoimmunotherapy of B-cell lymphoma. Mol. Oncol. 2016, 10, 1404–1414. [Google Scholar] [CrossRef] [PubMed]
  155. Zhang, Y.; Liu, Z.; Gao, C.; Bian, H.; Ma, Y.; Jing, F.; Zhao, X. Role of Rituximab in Treatment of Patients with Primary Central Nervous System Lymphoma (PCNSL): A Systematic Review and Meta-Analysis. Clin. Lymphoma Myeloma Leuk. 2023, 23, 733–741. [Google Scholar] [CrossRef]
  156. Narsinh, K.H.; Perez, E.; Haddad, A.F.; Young, J.S.; Savastano, L.; Villanueva-Meyer, J.E.; Winkler, E.; de Groot, J. Strategies to Improve Drug Delivery Across the Blood-Brain Barrier for Glioblastoma. Curr. Neurol. Neurosci. Rep. 2024, 24, 123–139. [Google Scholar] [CrossRef]
  157. Teleanu, R.I.; Preda, M.D.; Niculescu, A.G.; Vladâcenco, O.; Radu, C.I.; Grumezescu, A.M.; Teleanu, D.M. Current Strategies to Enhance Delivery of Drugs across the Blood-Brain Barrier. Pharmaceutics 2022, 14, 987. [Google Scholar] [CrossRef]
  158. Schroeder, H.W., Jr.; Cavacini, L. Structure and function of immunoglobulins. J. Allergy Clin. Immunol. 2010, 125, S41–S52. [Google Scholar] [CrossRef]
  159. Goulet, D.R.; Atkins, W.M. Considerations for the Design of Antibody-Based Therapeutics. J. Pharm. Sci. 2020, 109, 74–103. [Google Scholar] [CrossRef]
  160. Cheever, M.A.; Allison, J.P.; Ferris, A.S.; Finn, O.J.; Hastings, B.M.; Hecht, T.T.; Mellman, I.; Prindiville, S.A.; Viner, J.L.; Weiner, L.M.; et al. The prioritization of cancer antigens: A national cancer institute pilot project for the acceleration of translational research. Clin. Cancer Res. 2009, 15, 5323–5337. [Google Scholar] [CrossRef]
  161. Sethi, B.; Kumar, V.; Mahato, K.; Coulter, D.W.; Mahato, R.I. Recent advances in drug delivery and targeting to the brain. J. Control. Release 2022, 350, 668–687. [Google Scholar] [CrossRef]
  162. Wu, D.; Chen, Q.; Chen, X.; Han, F.; Chen, Z.; Wang, Y. The blood-brain barrier: Structure, regulation, and drug delivery. Signal Transduct. Target. Ther. 2023, 8, 217. [Google Scholar] [CrossRef] [PubMed]
  163. Chu, L.; Sun, Y.; Tang, X.; Duan, X.; Zhao, Y.; Xia, H.; Xu, L.; Zhang, P.; Sun, K.; Yang, G.; et al. The Tumor-Derived Exosomes Enhanced Bevacizumab across the Blood-Brain Barrier for Antiangiogenesis Therapy against Glioblastoma. Mol. Pharm. 2025, 22, 972–983. [Google Scholar] [CrossRef]
  164. Wen, J.; Wu, D.; Qin, M.; Liu, C.; Wang, L.; Xu, D.; Vinters, H.V.; Liu, Y.; Kranz, E.; Guan, X.; et al. Sustained delivery and molecular targeting of a therapeutic monoclonal antibody to metastases in the central nervous system of mice. Nat. Biomed. Eng. 2019, 3, 706–716. [Google Scholar] [CrossRef] [PubMed]
  165. Wei, D.; Tao, Z.; Shi, Q.; Wang, L.; Liu, L.; She, T.; Yi, Q.; Wen, X.; Liu, L.; Li, S.; et al. Selective Photokilling of Colorectal Tumors by Near-Infrared Photoimmunotherapy with a GPA33-Targeted Single-Chain Antibody Variable Fragment Conjugate. Mol. Pharm. 2020, 17, 2508–2517. [Google Scholar] [CrossRef] [PubMed]
  166. Zhao, Y.; Li, F.; Mao, C.; Ming, X. Multiarm Nanoconjugates for Cancer Cell-Targeted Delivery of Photosensitizers. Mol. Pharm. 2018, 15, 2559–2569. [Google Scholar] [CrossRef]
  167. Yamaguchi, H.; Suzuki, T.; Okada, Y.; Ono, J.; Sano, H.; Banba, A.; Sakata, H.; Ishikawa, A.; Morita, T. Near-Infrared Photoimmunotherapy Using a Protein Mimetic for EGFR-Positive Salivary Gland Cancer. Int. J. Mol. Sci. 2024, 25, 3233. [Google Scholar] [CrossRef]
  168. Yamaguchi, H.; On, J.; Morita, T.; Suzuki, T.; Okada, Y.; Ono, J.; Evdokiou, A. Combination of Near-Infrared Photoimmunotherapy Using Trastuzumab and Small Protein Mimetic for HER2-Positive Breast Cancer. Int. J. Mol. Sci. 2021, 22, 12213. [Google Scholar] [CrossRef]
  169. Watanabe, R.; Hanaoka, H.; Sato, K.; Nagaya, T.; Harada, T.; Mitsunaga, M.; Kim, I.; Paik, C.H.; Wu, A.M.; Choyke, P.L.; et al. Photoimmunotherapy targeting prostate-specific membrane antigen: Are antibody fragments as effective as antibodies? J. Nucl. Med. 2015, 56, 140–144. [Google Scholar] [CrossRef]
  170. 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]
  171. Ogawa, M.; Tomita, Y.; Nakamura, Y.; Lee, M.J.; Lee, S.; Tomita, S.; Nagaya, T.; Sato, K.; Yamauchi, T.; Iwai, H.; et al. Immunogenic cancer cell death selectively induced by near infrared photoimmunotherapy initiates host tumor immunity. Oncotarget 2017, 8, 10425–10436. [Google Scholar] [CrossRef]
  172. Mączyńska, J.; Da Pieve, C.; Burley, T.A.; Raes, F.; Shah, A.; Saczko, J.; Harrington, K.J.; Kramer-Marek, G. Immunomodulatory activity of IR700-labelled affibody targeting HER2. Cell Death Dis. 2020, 11, 886. [Google Scholar] [CrossRef] [PubMed]
  173. Nakajima, K.; Ogawa, M. Phototoxicity in near-infrared photoimmunotherapy is influenced by the subcellular localization of antibody-IR700. Photodiagnosis Photodyn. Ther. 2020, 31, 101926. [Google Scholar] [CrossRef]
  174. Yang, J.K.; Kwon, H.; Kim, S. Recent advances in light-triggered cancer immunotherapy. J. Mater. Chem. B 2024, 12, 2650–2669. [Google Scholar] [CrossRef] [PubMed]
  175. Yamaguchi-Takezawa, H.; Suzuki, T.; Banba, A.; Shibata, S.; Sakata, H.; Ishikawa, A.; Morita, T. Near-infrared photoimmunotherapy using a small protein mimetic for brain metastasis of HER2-positive breast cancer. J. Immunother. Cancer 2024, 12, A15–A16. [Google Scholar] [CrossRef]
  176. Kyrysyuk, O.; Wucherpfennig, K.W. Designing Cancer Immunotherapies That Engage T Cells and NK Cells. Annu. Rev. Immunol. 2023, 41, 17–38. [Google Scholar] [CrossRef]
  177. Jiang, Q.; Huang, J.; Zhang, B.; Li, X.; Chen, X.; Cui, B.; Li, S.; Guo, G. Efficacy and Safety of Anti-PD1/PDL1 in Advanced Biliary Tract Cancer: A Systematic Review and Meta-Analysis. Front. Immunol. 2022, 13, 801909. [Google Scholar] [CrossRef]
  178. Miliotou, A.N.; Papadopoulou, L.C. CAR T-cell Therapy: A New Era in Cancer Immunotherapy. Curr. Pharm. Biotechnol. 2018, 19, 5–18. [Google Scholar] [CrossRef]
  179. Caspi, R.R. Immunotherapy of autoimmunity and cancer: The penalty for success. Nat. Rev. Immunol. 2008, 8, 970–976. [Google Scholar] [CrossRef] [PubMed]
  180. Okada, R.; Maruoka, Y.; Furusawa, A.; Inagaki, F.; Nagaya, T.; Fujimura, D.; Choyke, P.L.; Kobayashi, H. The Effect of Antibody Fragments on CD25 Targeted Regulatory T Cell Near-Infrared Photoimmunotherapy. Bioconjugate Chem. 2019, 30, 2624–2633. [Google Scholar] [CrossRef]
  181. Ito, M.; Komai, K.; Mise-Omata, S.; Iizuka-Koga, M.; Noguchi, Y.; Kondo, T.; Sakai, R.; Matsuo, K.; Nakayama, T.; Yoshie, O.; et al. Brain regulatory T cells suppress astrogliosis and potentiate neurological recovery. Nature 2019, 565, 246–250. [Google Scholar] [CrossRef]
  182. Cugurra, A.; Mamuladze, T.; Rustenhoven, J.; Dykstra, T.; Beroshvili, G.; Greenberg, Z.J.; Baker, W.; Papadopoulos, Z.; Drieu, A.; Blackburn, S.; et al. Skull and vertebral bone marrow are myeloid cell reservoirs for the meninges and CNS parenchyma. Science 2021, 373, eabf7844. [Google Scholar] [CrossRef]
  183. Nakajima, K.; Sugikawa, A.; Yasui, H.; Higashikawa, K.; Suzuki, C.; Natsume, T.; Suzuki, M.; Takakura, H.; Tomita, M.; Takahashi, S.; et al. In vivo imaging of acute physiological responses after treatment of cancer with near-infrared photoimmunotherapy. Mol. Imaging Biol. 2023, 25, 648–658. [Google Scholar] [CrossRef] [PubMed]
  184. Ma, G.; Gordon, M.; Lapetoda, J.; Lozano, A.; Amantea, C.M.; Osada, T.; Thorne, A.H. Reduction in photoimmunotherapy-induced edema with COX-2 inhibition: Combatting clinically relevant adverse events without compromising efficacy. In Proceedings of the American Association for Cancer Research, San Diego, CA, USA, 5–10 April 2024. [Google Scholar]
  185. Layard Horsfall, H.; Mohan, M.; Devi, B.I.; Adeleye, A.O.; Shukla, D.P.; Bhat, D.; Khan, M.; Clark, D.J.; Chari, A.; Servadei, F.; et al. Hinge/floating craniotomy as an alternative technique for cerebral decompression: A scoping review. Neurosurg. Rev. 2020, 43, 1493–1507. [Google Scholar] [CrossRef]
  186. Kumar Sengupta, S. Expansive Cranioplasty a Single Stage Surgery Avoiding Cranioplasty at a Later Date Following Decompressive Hemicraniectomy. World Neurosurg. 2019, 128, 631. [Google Scholar] [CrossRef]
  187. Wang, J.; Ren, Y.; Zhou, L.-J.; Kan, L.-D.; Fan, H.; Fang, H.-M. Glycerol Infusion Versus Mannitol for Cerebral Edema: A Systematic Review and Meta-analysis. Clin. Ther. 2021, 43, 637–649. [Google Scholar] [CrossRef] [PubMed]
  188. Vredenburgh, J.J.; Cloughesy, T.; Samant, M.; Prados, M.; Wen, P.Y.; Mikkelsen, T.; Schiff, D.; Abrey, L.E.; Yung, W.K.; Paleologos, N.; et al. Corticosteroid use in patients with glioblastoma at first or second relapse treated with bevacizumab in the BRAIN study. Oncologist 2010, 15, 1329–1334. [Google Scholar] [CrossRef] [PubMed]
  189. Okamoto, I.; Okada, T.; Tokashiki, K.; Tsukahara, K. Photoimmunotherapy for Managing Recurrent Laryngeal Cancer Cervical Lesions: A Case Report. Case Rep. Oncol. 2022, 15, 34–39. [Google Scholar] [CrossRef]
  190. Omura, G.; Honma, Y.; Matsumoto, Y.; Shinozaki, T.; Itoyama, M.; Eguchi, K.; Sakai, T.; Yokoyama, K.; Watanabe, T.; Ohara, A.; et al. Transnasal photoimmunotherapy with cetuximab sarotalocan sodium: Outcomes on the local recurrence of nasopharyngeal squamous cell carcinoma. Auris Nasus Larynx 2023, 50, 641–645. [Google Scholar] [CrossRef]
Figure 1. Mechanism of NIR-PIT: NIR-PIT utilizes IRDye700DX–antibody conjugates. When these conjugates bind to cancer cell receptors and are activated by 690 nm near-infrared light, IR700 undergoes photochemical reactions that convert it from hydrophilic to hydrophobic. This transformation leads to molecular aggregation, which disrupts cell membrane integrity and induces cell death. Solid black arrows indicate IR700Dye in each conjugate, while dotted black arrows indicate H2O entering the cancer cells.
Figure 1. Mechanism of NIR-PIT: NIR-PIT utilizes IRDye700DX–antibody conjugates. When these conjugates bind to cancer cell receptors and are activated by 690 nm near-infrared light, IR700 undergoes photochemical reactions that convert it from hydrophilic to hydrophobic. This transformation leads to molecular aggregation, which disrupts cell membrane integrity and induces cell death. Solid black arrows indicate IR700Dye in each conjugate, while dotted black arrows indicate H2O entering the cancer cells.
Pharmaceuticals 18 00751 g001
Figure 2. (a) The fluorescence intensity of the head increased dramatically immediately after the injection of the EGFR Affibody–IR700Dye conjugate. (b) Brain collection and fluorescence imaging were performed 1 h after i.v. injection of the EGFR Affibody–IR700 conjugate. (c) Fluorescent images of IR700 and hematoxylin/eosin staining after the brain was fixed overnight. Fluorescence accumulation is observed at the tumor site. The yellow arrows show the progression of the experimental timeline from left to right.
Figure 2. (a) The fluorescence intensity of the head increased dramatically immediately after the injection of the EGFR Affibody–IR700Dye conjugate. (b) Brain collection and fluorescence imaging were performed 1 h after i.v. injection of the EGFR Affibody–IR700 conjugate. (c) Fluorescent images of IR700 and hematoxylin/eosin staining after the brain was fixed overnight. Fluorescence accumulation is observed at the tumor site. The yellow arrows show the progression of the experimental timeline from left to right.
Pharmaceuticals 18 00751 g002
Table 1. The differences between NIR-PIT and PDT.
Table 1. The differences between NIR-PIT and PDT.
CharacteristicsNIR-PITPDT
PhotosensitizerIR700 conjugated to antibodiesTalaporfin sodium
Excitation wavelength690 nm664 nm
Penetration depthApproximately 2 cm [24]<10 mm [29,30]
Cell death mechanismDisruption of cell membranesApoptosis via ROS generation
SpecificityHigh (targets only cells expressing specific antigens) Moderate (preferential accumulation in tumors)
Disclaimer/Publisher’s Note: The statements, opinions and data contained in all publications are solely those of the individual author(s) and contributor(s) and not of MDPI and/or the editor(s). MDPI and/or the editor(s) disclaim responsibility for any injury to people or property resulting from any ideas, methods, instructions or products referred to in the content.

Share and Cite

MDPI and ACS Style

Yamaguchi, H.; Okada, M.; Otani, T.; On, J.; Shibuma, S.; Takino, T.; Watanabe, J.; Tsukamoto, Y.; Ogura, R.; Oishi, M.; et al. Near-Infrared Photoimmunotherapy in Brain Tumors—An Unexplored Frontier. Pharmaceuticals 2025, 18, 751. https://doi.org/10.3390/ph18050751

AMA Style

Yamaguchi H, Okada M, Otani T, On J, Shibuma S, Takino T, Watanabe J, Tsukamoto Y, Ogura R, Oishi M, et al. Near-Infrared Photoimmunotherapy in Brain Tumors—An Unexplored Frontier. Pharmaceuticals. 2025; 18(5):751. https://doi.org/10.3390/ph18050751

Chicago/Turabian Style

Yamaguchi, Haruka, Masayasu Okada, Takuya Otani, Jotaro On, Satoshi Shibuma, Toru Takino, Jun Watanabe, Yoshihiro Tsukamoto, Ryosuke Ogura, Makoto Oishi, and et al. 2025. "Near-Infrared Photoimmunotherapy in Brain Tumors—An Unexplored Frontier" Pharmaceuticals 18, no. 5: 751. https://doi.org/10.3390/ph18050751

APA Style

Yamaguchi, H., Okada, M., Otani, T., On, J., Shibuma, S., Takino, T., Watanabe, J., Tsukamoto, Y., Ogura, R., Oishi, M., Suzuki, T., Ishikawa, A., Sakata, H., & Natsumeda, M. (2025). Near-Infrared Photoimmunotherapy in Brain Tumors—An Unexplored Frontier. Pharmaceuticals, 18(5), 751. https://doi.org/10.3390/ph18050751

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